You are on page 1of 37

Neuromodulation: Technology at the Neural Interface

Received: March 4, 2016 Revised: May 23, 2016 Accepted: May 23, 2016

(onlinelibrary.wiley.com) DOI: 10.1111/ner.12538

The Polyanalgesic Consensus Conference


(PACC): Recommendations on Intrathecal Drug
Infusion Systems Best Practices and Guidelines
Timothy R. Deer, MD*; Jason E. Pope, MD†; Salim M. Hayek, MD, PhD‡;
Anjum Bux, MD§; Eric Buchser, MD¶; Sam Eldabe, MD**;
Jose A. De Andre s, MD, PhD, EDRA††; Michael Erdek, MD‡‡;
Dennis Patin, MD§§; Jay S. Grider, PhD, MBA¶¶; Daniel M. Doleys, PhD***;
Marilyn S. Jacobs, PhD†††; Tony L. Yaksh, PhD‡‡‡;
Lawrence Poree, MD, PhD§§§; Mark S. Wallace, MD¶¶¶; Joshua Prager, MD****;
Richard Rauck, MD††††; Oscar DeLeon, MD‡‡‡‡; Sudhir Diwan, MD§§§§;
Steven M. Falowski, MD¶¶¶¶; Helena M. Gazelka, MD*****;
Philip Kim, MD†††††‡‡‡‡‡; Michael Leong, MD§§§§§;
Robert M. Levy, MD, PhD¶¶¶¶¶; Gladstone McDowell II, MD******;
Porter McRoberts, MD††††††; Ramana Naidu, MD‡‡‡‡‡‡;
Samir Narouze, MD, PhD§§§§§§; Christophe Perruchoud, MD¶¶¶¶¶¶;
Steven M. Rosen, MD*******; William S. Rosenberg, MD†††††††;
Michael Saulino, MD, PhD‡‡‡‡‡‡‡; Peter Staats, MD§§§§§§§¶¶¶¶¶¶¶;
Lisa J. Stearns, MD********; Dean Willis, MD††††††††; Elliot Krames, MD‡‡‡‡‡‡‡‡;
Marc Huntoon, MD§§§§§§§§; Nagy Mekhail, MD, PhD¶¶¶¶¶¶¶¶
Introduction: Pain treatment is best performed when a patient-centric, safety-based philosophy is used to determine an algorithmic process to
guide care. Since 2007, the International Neuromodulation Society has organized a group of experts to evaluate evidence and create a Polyanalge-
sic Consensus Conference (PACC) to guide practice.
Methods: The current PACC update was designed to address the deficiencies and innovations emerging since the previous PACC publication of
2012. An extensive literature search identified publications between January 15, 2007 and November 22, 2015 and authors contributed additional
relevant sources. After reviewing the literature, the panel convened to determine evidence levels and degrees of recommendations for intrathecal
therapy. This meeting served as the basis for consensus development, which was ranked as strong, moderate or weak. Algorithms were developed
for intrathecal medication choices to treat nociceptive and neuropathic pain for patients with cancer, terminal illness, and noncancer pain, with
either localized or diffuse pain.
Results: The PACC has developed an algorithmic process for several aspects of intrathecal drug delivery to promote safe and efficacious
evidence-based care. Consensus opinion, based on expertise, was used to fill gaps in evidence. Thirty-one consensus points emerged from the
panel considerations.
Conclusion: New algorithms and guidance have been established to improve care with the use of intrathecal drug delivery.

Address correspondence to: Timothy R. Deer, MD, President and CEO, The Center for Pain Relief, Inc., 400 Court Street, Ste 100, Charleston, WV, USA. Email: DocTDeer@
aol.com

* Center for Pain Relief, Charleston, WV, USA;



Summit Pain Alliance, Santa Rosa, CA, USA;

Case Western Reserve University, Cleveland, OH, USA;
§
Anesthesia and Chronic Pain Management, Ephraim McDowell Regional Medical Center, Danville, KY, USA;

Anaesthesia and Pain Management Department, EHC Hosptial, Morges, and CHUV University Hospital, Lausanne, Switzerland;
** The James Cook University Hospital, Middlesbrough, UK;
††
Valencia School of Medicine, Hospital General Universitario, Valencia, Spain;
‡‡
Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA;
§§
University of Miami Health System, Miami, FL, USA;
¶¶
University of Kentucky College of Medicine, UK HealthCare Pain Services, Lexington, KY, USA;
*** Doleys Clinic, Birmingham, AL, USA;
†††
California Pain Medicine Center, Los Angeles, CA, USA;
96

‡‡‡
Anesthesiology and Pharmacology, University of California, San Diego, CA, USA;

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Keywords: Chronic pain, consensus, fixed rate pump, intrathecal drug delivery, neuropathic pain, nonmalignant pain, opioid,
programmable pump, psychological evaluation, safety
Conflict of Interest: Dr. Deer is a consultant for St. Jude Medical Inc., Medtronic Inc., Bioness Inc., Vertos Medical Inc., Nevro Corp., Flowonix Medical Inc.,
Axonics Ltd., Ethos, Spinal Therapeutics, Saluda Medical Pty. Ltd., and Nuvectra Corp. He serves on the Advisory Board of St. Jude Medical Inc., Medtronic
Inc., Bioness Inc., Nevro Corp., Flowonix Medical Inc., Jazz Pharmaceuticals PLC, and Axonics Ltd. He is also a speaker for Jazz Pharmaceuticals PLC, and has
stock options in Bioness Inc., Vertos Medical Inc., Axonics Ltd., Spinal Therapeutics, Saluda Medical Pty. Ltd., and Nuvectra Corp. Dr. Pope is a consultant for
Medtronic Inc., St. Jude Medical Inc., Jazz Pharmaceuticals PLC, Nevro Corp., Nuvectra Corp., and Flowonix Medical Inc. Dr. Hayek is a consultant for Flowo-
nix Medical Inc. and Mallinckrodt Pharmaceuticals. Dr. Bux receives speaking honoraria from Medtronic Inc., and Jazz Pharmaceuticals PLC. Dr. Buchser is a
consultant and researcher for Medtronic Inc. Dr. Eldabe is a consultant for Medtronic Inc. Dr. De Andr es has no conflicts of interest to report. Dr. Erdek has
no conflicts of interest to report. Dr. Patin is a consultant for Medtronic Inc. Dr. Grider is a consultant for Interlink Spine, and is a speaker for Medtronic Inc.
Dr. Doleys is a speaker for Medtronic Inc. and Kal eo Pharma/Evzio. Dr. Jacobs has no conflicts of interest to report. Dr. Yaksh is a consultant for Adynxx, and
a researcher for Medtronic Inc., Flowonix Medical Inc., and Jazz Pharmaceuticals PLC. Dr. Poree is on the Medtronic Inc. Safety Advisory Panel. He is a consul-
tant for Medtronic Inc., Nalu Medical Inc., Stimwave Technology Ltd., St. Jude Medical Inc., and Mallinckrodt Pharmaceuticals. Dr. Wallace is a consultant for
Jazz Pharmaceuticals PLC and the Alfred Mann Foundation. Dr. Prager is a speaker for Insys Therapeutics and a consultant for Medtronic Inc. Dr. Rauck
advises and conducts research for Jazz Pharmaceuticals PLC and Medtronic Inc. He is also a speaker for Jazz Pharmaceuticals PLC. Dr. De Leon-Casasola is a
consultant for Mallinckrodt Pharmaceuticals, Purdue Pharma LP, and Depomed Inc. Dr. Diwan has no conflicts of interest to report. Dr. Falowski is a consul-
tant, researcher, and speaker for Medtronic Inc. and St. Jude Medical Inc. He is a consultant and speaker for Nevro Corp., and a researcher for Saluda
Medical Pty. Ltd. Dr. Gazelka has no conflicts of interest to report. Dr. Kim is a consultant and speaker for Medtronic Inc. and Jazz Pharmaceuticals PLC, and
is a consultant for Biotronic NeuroNetwork. Dr. Leong is a consultant for Boston Scientific Corp., Jazz Pharmaceuticals PLC, and Sorrento Therapeutics. Dr.
Levy is a minority shareholder in Spinal Modulation Inc., Bioness Inc., Vertos Medical Inc., and Nevro Corp. He is a consultant for Medtronic Inc., St. Jude
Medical Inc., Spinal Modulation Inc., Nevro Corp., Saluda Medical Pty. Ltd., and Flowonix Medical Inc. Dr. McDowell is an advisor and consultant for Med-
tronic Inc., Jazz Pharmaceuticals PLC, and Flowonix Medical Inc. Dr. McRoberts is an advisor for St. Jude Medical Inc., Medtronic Inc., Flowonix Medical Inc.,
Nalu Medical Inc., Vertiflex Inc., Boston Scientific Corp., Bioness Inc, SPR, Interlink Spine and OrthoSensor. He is a researcher for Mesoblast, Medtronic Inc.,
Vertiflex Inc., and Mainstay Medical. He is a speaker for Nevro Corp., and St. Jude Medical Inc. Dr. Naidu is a consultant and speaker for Halyard Health. Dr.
Narouze is a consultant for St. Jude Medical Inc. Dr. Perruchoud is a consultant, researcher, and speaker for Medtronic Inc. He is a consultant and researcher
for Spinal Modulation Inc., and is a consultant for St. Jude Medical Inc. Dr. Rosen is a consultant and researcher for Flowonix Medical Inc. and Saluda Medical
Pty. Ltd., and a researcher for Nevro Corp. Dr. Rosenberg is a consultant and speaker for Medtronic Inc. and St. Jude Medical Inc. Dr. Saulino is a researcher
and speaker for Medtronic Inc., Mallinckrodt Pharmaceuticals and Jazz Pharmaceuticals PLC. Dr. Staats is a consultant for ElectroCore Medical LLC, St. Jude
Medical Inc., Boston Scientific Corp., and Medtronic Inc. He does research for Bioness Inc., Spinal Modulation Inc., Boston Scientific Corp., Vertos Medical
Inc., St. Jude Medical Inc., Saluda Medical Pty. Ltd., Gr€
uenthal GmbH, and ElectroCore Medical LLC. He receives product royalty and licensing fees from Neu-
rogesX Inc., and has relevant holdings in ElectroCore Medical LLC. Dr. Stearns is a consultant for Medtronic Inc., Spinal Modulation Inc., Pfizer Inc., and the
Alfred Mann Foundation. Dr. Willis has no conflicts to disclose. Dr. Krames has no conflicts to report. Dr. Huntoon is a researcher for CNS Therapeutics and a
part-time employee of Regional Anesthesia and Pain Medicine. Dr. Mekhail is an advisor for Flowonix Medical Inc., Boston Scientific Corp. and Saluda Medical
Pty. Ltd. He is a researcher for Mesoblast Ltd., Halyard Health, CNS Therapeutics, Axsome Therapeutics and Stimwave Technology Ltd.

§§§
Pain Clinic of Monterey Bay, University of California at San Francisco, San Francisco, CA, USA;
¶¶¶
Department of Anesthesia, UC San Diego, San Diego, CA, USA;
**** Center for the Rehabilitation Pain Syndromes (CRPS) at UCLA Medical Plaza, Los Angeles, CA, USA;
††††
Carolina Pain Institute, Wake Forest Baptist Health, Winston-Salem, NC, USA;
‡‡‡‡
Roswell Park Cancer Institute, SUNY, Buffalo, NY, USA;
§§§§
Manhattan Spine and Pain Medicine, Lenox Hill Hospital, New York, NY, USA;
¶¶¶¶
St. Luke’s University Health Network, Bethlehem, PA, USA;
***** Mayo Clinic, Rochester, MN, USA;
†††††
Bryn Mawr Hospital, Bryn Mawr, PA, USA;
‡‡‡‡‡
Christiana Hospital, Newark, DE, USA;
§§§§§
Stanford University, Palo Alto, CA, USA;
¶¶¶¶¶
Marcus Neuroscience Institute, Boca Raton, FL, USA;
****** Integrated Pain Solutions, Columbus, OH, USA;
††††††
Holy Cross Hospital, Fort Lauderdale, Florida, USA;
‡‡‡‡‡‡
San Francisco Medical Center, University of California, San Francisco, CA, USA;
§§§§§§
Summa Western Reserve Hospital, Cuyahoga Falls, OH, USA;
¶¶¶¶¶¶
H^opital de Zone Morges, Morges, Switzerland;
******* Fox Chase Pain Management Associates PC, Doylestown, PA, USA;
†††††††
Center for Relief of Pain, Kansas City, MO, USA;
‡‡‡‡‡‡‡
MossRehab, Elkins Park, PA, USA;
§§§§§§§
Premier Pain Management Centers, Shrewsbury, NJ, USA;
¶¶¶¶¶¶¶
Johns Hopkins University, Baltimore, MD, USA;
******** Center for Pain and Supportive Care, Phoenix, AZ, USA;
††††††††
Alabama Pain Center, Huntsville, AL, USA;
‡‡‡‡‡‡‡‡
Pacific Pain Treatment Center (ret.), San Francisco, CA, USA;
§§§§§§§§
Vanderbilt University Medical Center, Nashville, TN, USA; and
¶¶¶¶¶¶¶¶
Cleveland Clinic, Cleveland, OH, USA

For more information on author guidelines, an explanation of our peer review process, and conflict of interest informed consent policies, please go to http://www.wiley.
com/WileyCDA/Section/id-301854.html
97

[Correction added on 19 January 2017, after first online publication: the name of the third author has been corrected from “Salim Hayek” to “Salim M. Hayek”.]

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

INTRODUCTION AND RATIONALE tailor algorithmic approaches to patient-specific characteristics that


likely impact IT therapy (2,6–9). A number of clinical factors play
The use of intraspinal (intrathecal [IT]) infusion of analgesic medica- important roles in shaping specific IT interventions and medication
tions to treat patients with chronic refractory pain has increased since choices. These factors have been previously described (10), and
its inception in the 1980s, and the need for clinical and outcomes include patient diagnoses and expected survival time (11), patient
research in IT therapy is ongoing. New IT devices have been recently age (12,13), previous exposure to opioids (13–15), location of pain
introduced, along with novel chronic infusion strategies (1). Thus far, (diffuse vs. localized), type of pain (nociceptive, neuropathic, or
research has not kept pace with the growing need for innovative IT mixed), the physiochemical properties of lipid solubility of the IT
pain management, and clinical care and decision making have largely medications employed (16,17), cerebrospinal fluid (CSF) flow
relied on best evidence and expert opinion (2). Therefore, a consen- dynamics and pharmacokinetics (18,19), IT catheter location (20),
sus opinion is needed to identify the current research and address pump and catheter characteristics, kinetics of the IT infusate (20),
deficiencies in the data. Furthermore, as new IT therapies become and psychological status (21–23) of the patient with chronic pain. A
available, the need to refine patient selection and pain care algo- detailed discussion of these factors will be entertained in the follow-
rithms is required (3). With more than 80% of IT therapy in the United ing sections. Each component to be considered requires careful and
States employed as off-label (4,5), there is a continuing need to help deliberate attention. Interestingly, unpublished industry reports
navigate careful decision-making surrounding IT therapy. suggest that many practicing physicians in the United States and
Europe do not follow the PACC guidance of 2012 (24). Physicians
Consensus Point 1. An update of the best practices of IT drug and scientists have been impacted by the advice given in this
delivery is needed due to many changes in patient care since the process, with nearly 100 citations of the 2012 guidance. The effort
last version of this living document. of the 2016 PACC is to provide recommendations based on
Consensus Point 2. Evidence for practice guidance has been evidence and consensus and to continue to disseminate best
improved since 2012, but additional expert guidance is needed to practice insights to practicing physicians worldwide.
fill the current gaps in clinical practice. Consensus Point 3. The 2016 PACC will continue the historical
The Polyanalgesic Consensus Conference (PACC) panel was goal of improving safety and efficacy of the global use of IT therapies.
formed to improve the safety and efficacy of IT therapy. PACC was
initiated in 2000 to address the research gaps and review the existing
data regarding IT therapy (2). This expert panel was composed of METHODS
health care providers within the field of IT therapy. The panel devel-
oped an IT drug selection algorithm on the basis of best evidence The PACC of 2016 was designed to address the deficiencies and
and expert opinion, prepared supplemental reports that included a innovations emerging since the previous PACC of 2012 regarding IT
relevant literature review, reported on results from a survey of peers therapy. Participants were chosen based on an executive panel from
engaged in IT therapy, and described future directions in the field of the International Neuromodulation Society (INS), with participants
IT therapy. The PACC panel reconvened in 2003 (6) and 2007 (7) to from previous PACC guidelines automatically nominated. Other
evaluate the most up-to-date literature and to update the algorithm nominations were made by board members based on a needs
for intraspinal drug selection. In 2011, to formulate consensus opin- assessment of topics to be addressed. All participants were identi-
ions on critical issues involving IT therapy and identify areas for fied to have an area of needed expertise, which could include exten-
future research in the field, the PACC panel again convened, adding sive experience in IT drug device management, basic science
a supplement on IT granuloma and describing new insights on rec- research, clinical studies, or expertise in evidence assessment or pub-
ommended maximal concentrations and daily doses of IT agents (8). lication. Invitations were subsequently sent to potential participants
Since then, renewed interest regarding noncancer pain management and accepted prior to formal engagement. Meetings were held peri-
employing IT agents has resurfaced, along with new interests on odically during the composition and drafting of the manuscript,
infusion schema. This present update focuses on redefining patient- with meetings to rank evidence and develop consensus surrounding
centric trialing strategies and the recommended observation period IT therapy, as defined below. The authorship publication standards
for trialing, algorithmic care for the near end-of-life patient, and the outlined by the journal Neuromodulation and Wiley Publishing gov-
importance of catheter position congruent with regional location of erned the working consensus group.
the pain. The overarching theme of this present PACC update is to
continue to modify and adapt this living document. Literature Search Methods
A broad literature search was conducted to identify preclinical
Brief History of PACC and clinical data on IT therapy published from January 15, 2007,
In spite of thorough discussions of various factors impacting through November 22, 2015. MEDLINEV R , BioMed CentralV
R , Current

patient responses to IT therapy, previous PACC versions did not Contents ConnectV R, EmbaseTM, International Pharmaceutical

Table 1. Hierarchy of Studies by the Type of Design (U.S. Preventive Services Task Force, Ref [25]).

Evidence level Study type

I At least one controlled and randomized clinical trial, properly designed


II-1 Well-designed, controlled, nonrandomized clinical trials
II-2 Cohort or case studies and well-designed controls, preferably multicenter
II-3 Multiple series compared over time, with or without intervention, and surprising results in noncontrolled experiences
III Clinical experience-based opinions, descriptive studies, clinical observations or reports of expert committees.
98

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Table 2. Meaning of Recommendation Degrees (U.S. Preventive Services Task Force, Ref [25]).

Degree of recommendation Meaning

A Extremely recommendable (good evidence that the measure is effective and benefits outweigh the harms)
B Recommendable (at least, moderate evidence that the measure is effective and benefits exceed harms)
C Neither recommendable nor inadvisable (at least moderate evidence that the measure is effective, but benefits are
similar to harms and a general recommendation cannot be justified)
D Inadvisable (at least moderate evidence that the measure is ineffective or that the harms exceed the benefits)
I Insufficient, low quality or contradictory evidence; the balance between benefit and harms cannot be determined.

AbstractsVR , and Web of ScienceVR , Google Scholar, and Pubmed data review. These searches yielded 391 articles, which were examined
bases were searched for publications on a range of medications that for relevance to the IT treatment of chronic pain. Google Scholar
are either currently in use or potentially useful for the IT treatment was again searched for recent relevant information regarding IT
of chronic pain. Search terms included “intrathecal, intraspinal, mor- therapy for chronic pain, and additional literature considered by
phine, fentanyl, sufentanil, methadone, adenosine, hydromorphone, panel members to be relevant to this new consensus paper was
meperidine, gabapentin, baclofen, ketorolac, midazolam, neostig- reviewed. Wherever pertinent, proposed mechanisms of action for
mine, octreotide, ziconotide, ropivacaine, dexmedetomidine, cloni- the particular drug class are provided, along with a summary of pre-
dine, bupivacaine, and lidocaine.” Each author performed clinical studies, followed by critical review. Literature published
independent literature searches and the information was cross- before the dates stated above is cited when relevant. After review-
referenced and compiled for evidence analysis and consensus ing the literature, the PACC panel convened to develop

99

Figure 1. Contributor evidence assessment.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

opinions and recommendations offered are not intended to pro-


Table 3. Strength of Consensus.
mote off-label uses of medications and devices. Additionally, these
recommendations should not be construed as a standard of care.
Strength of consensus Definition*
We will explore an evidence-based algorithm of pain care, patient
Strong >80% consensus selection, drug selection, trialing strategies, implantation, and con-
Moderate 50–79% consensus centration and dosing. Physicians should consult their national
Weak <49% consensus approval processes when making clinical decisions.
*Quorum defined as 80% of participants available for vote. It is important to address the conflicting nature of evidence and
the need for consensus. Evidence and consensus are not mutually
exclusive, which may be the perception at first glance. Evidence
assessment, regardless of the strength, needs interpretation for clini-
recommendations for IT analgesia. Supporting literature is included cal application whenever used.
following these recommendations and discussions of the panel.
RECOMMENDATIONS OF PACC 2016
Evidence-Based Analysis vs. Opinion
Similar to the Neuromodulation Appropriateness Consensus Con- In this manuscript, we will explore the evidence-weighted and
ference (NACC) of the International Neuromodulation Society publi- consensus recommendations of the PACC regarding the following
cation in 2014, and fostered from the previous PACC statements, the topics:
goal of this present PACC effort was to create a living document,
• Evidence assessment
with continued refreshment and evidence synthesis ongoing, as
• Pain care algorithms
appropriate. Unlike the PACC of 2012, the effort of the 2016 PACC
• Disease-specific indications and considerations
was to apply a validated evidence-ranking system, outlining man-
• Patient-selection considerations
agement and implementation of IT therapy. This effort was under-
• Medication-selection recommendations and considerations
scored by unpublished survey data suggesting poor adoption of the • Use context of neuropathic and nociceptive pain
previous proposed algorithms (personal communication with Med- • Recommended starting dosages
tronic plc. and Jazz Pharmaceuticals). It is clear, however, that IT • Variables affecting chronic intrathecal therapy
granuloma identification and management were significantly • Conclusions
impacted and improved on since the PACC of 2011 supplement was
published (24).

Evidence Ranking
EVIDENCE ASSESSMENT
The United States Preventative Services Task Force (USPSTF) creat-
It is generally regarded that IT therapy offers a reliable, accurate,
ed hierarchies of studies and degrees of recommendations based on safe, and efficacious treatment for both cancer and noncancer pain,
evidence rankings as outlined in Tables 1 and 2 (25). The PACC of as well as for end-of-life pain care. There have been multiple reviews
2016 has adopted these classifications, just as the NACC previously discussing the efficacy and safety of IT therapy (26–29). Recently, IT
adopted weighted recommendations for neurostimulation. therapy options came under scrutiny by the state of California, with
Authors of this manuscript were asked to complete reference success of continued access available only after a demonstration of
forms for their section’s assessment (Fig. 1). These forms were evidence was ruled favorable (30). A more thorough understanding
then reviewed by the executive committee of the working group of the pharmacokinetic properties of IT medications (20) and CSF
and averaged. They served as the basis for review and consensus flow dynamics within the IT space was the catalyst in creating better
development. The working group developed recommendations IT therapy strategies (31). Furthermore, as defined previously, there
based on evidence ranking, or consensus when evidence was is evidence to support its use (26), based on the USPSTF criteria for
lacking, followed by assigning consensus rankings. The consensus data ranking and evidence strength. USPSTF strength of evidence
determination was performed during in-person meetings or via for IT therapy was level II-3 for noncancer pain and level II-2 for can-
teleconference with a quorum of 80% of the contributing authors cer pain. A best practice article was written in 2014 by Prager et al.,
determining recommendation strength. Consensus rankings were providing evidence in support of IT therapy and the needed place-
outlined as strong, moderate, or weak based on agreement, as ment of IT therapy in the algorithm for cancer and noncancer pain
defined in Table 3. (27) (Table 4). The support for efficacy of IT opioid administration for
As with any guideline, this document serves as a recommendation the management of chronic noncancer pain comes largely from pro-
regarding implementation and management of IT therapy. The spective and retrospective noncontrolled trials. Ziconotide has been

Table 4. Recommendations for Evidence Assessment of Intrathecal Therapy by the PACC Using USPSTF Criteria.

Statement Evidence level Recommendation grade Consensus level

Intrathecal therapy should be utilized for active cancer-related pain with opioids. I A Strong
Intrathecal therapy should be utilized for active cancer-related pain with ziconotide. I A Strong
Intrathecal therapy should be utilized for noncancer-related pain with opioids. III B Strong
100

Intrathecal therapy should be utilized for noncancer-related pain with ziconotide. I A Strong

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Figure 2. Algorithm for placement within the pain care algorithm for noncancer or non-end-of-life pain. DRG, dorsal root ganglion; HF10, high frequency stimula-
tion; PNfS, peripheral nerve field stimulation; PNS, peripheral nerve stimulation; SCS, spinal cord stimulation. Green arrows indicate affirmation or positive response;
red arrows signify negative response.

robustly studied in three randomized, placebo-controlled trials, Algorithmic Treatment of Pain


demonstrating safety and efficacy for both cancer and noncancer The landscape of pain medicine and IT therapy has evolved (35).
pain (32–34). In a randomized controlled trial (RCT) in cancer pain, Neuromodulation literature supports that it is more cost effective,
which compared efficacy and side effects for IT delivery of opioids more efficacious, and no longer appropriate to position IT therapy
vs. systemic delivery via conservative management, IT delivery of as a salvage therapy (36). The choice of neuromodulation therapy
opioids was superior (11). rests on many factors, including the regional location of the pain,
At the present time, there is an ongoing investigative effort, type of pain, life expectancy, and malignancy, to name a few. Gener-
with FDA oversight, by Mallinckrodt Pharmaceuticals (St. Louis, ally, and most significantly, IT therapy is not salvage therapy after
MO) to move IT hydromorphone from a compounded to a failure of systemic high-dose opioid-based medicines. IT drug deliv-
branded, formally manufactured, FDA-approved product. This ery devices (IDDs) should be suggested for refractory pain, as recent-
endeavor involves two clinical trials. The first trial is a controlled, ly defined (37):
two-arm, parallel group, randomized withdrawal study followed
Pain is defined as refractory, regardless of etiology, when 1)
by an openlabel single-arm safety study of hydromorphone. Fur-
ther, critical evaluations regarding sustainability and cost effec- multiple evidence-based biomedical therapies used in a clini-
tiveness have been performed, with focus on sustainability and cally appropriate and acceptable fashion have failed to reach
safety. treatment goals that may include adequate pain reduction
and/or improvement in daily functioning or have resulted in
Consensus Point 4. The use of evidence ranking is a critical piece
intolerable adverse effects, and when 2) psychiatric disorders
of the formatting of the 2016 PACC. This is the first time this impor-
and psychosocial factors that could influence pain outcomes
tant point has been included in the PACC methods.
have been assessed and appropriately optimized.
Consensus Point 5. In areas where evidence is strong, peer-
reviewed references are noted for the PACC recommendation. A proposed algorithm has been described (27,38). A modified ver-
When evidence is weak or lacking, consensus opinion is used to sion of the algorithm is suggested here (Fig. 2).
make recommendations. Important decision-tree aspects of this algorithm deserve men-
tion. IT opioid infusion within the algorithm deserves special consid-
eration. Compared to systemic opioid delivery, IT opioids appear to
PAIN CARE ALGORITHM be safer based on mortality and morbidity data (39) and have fewer
diversion risks. From 2003 to 2013, opioid use disorder increased in
Careful consideration of patient selection is foundational for suc- the United States, significantly increasing mortality, with more than
cessful, sustainable patient care. The fact that no recommendations 16,000 deaths reported annually (40). This has led the Centers for
were made regarding patient survival and IT therapy, or anatomic Disease Control (CDC) to draft new guidelines for chronic opioid
region of pain, were deficiencies of the previous 2012 PACC. There- therapy for noncancer pain. The first recommendation of those
fore, the PACC of 2016 is presenting evidence and consensus-based guidelines stresses the need for nonpharmacological and nonopioid
recommendations regarding patient survival, disease process, and treatments before resorting to systemic opioids. Along with neuro-
medication usage for IT therapy. Attention was directed to the age stimulation, IT therapy should play an important role in the pain
of the patient, although the contributions of age are reflected in treatment continuum. The reported challenges associated with IT
dosage sustainability, which is addressed elsewhere in the therapy (41,42) have been increasingly mitigated by vigilance and
101

recommendations. mindful patient selection (43).

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

Figure 3. Pain care algorithm for cancer-related pain. DRG, dorsal root ganglion; HF10, high frequency stimulation; PNfS, peripheral nerve field stimulation; PNS,
peripheral nerve stimulation; SCS, spinal cord stimulation. Green arrows indicate affirmation or positive response; red arrows signify negative response.

As can be gleaned from Figure 2, and new to descriptions of algo- (52). From a historical perspective, IT coverage may be influenced by
rithmic advanced pain care, IT therapy now occupies the same line catheter position and medication selection, providing flexibility to
of management as neurostimulation, with important caveats. If the the clinician and the patient (8). Coverage of a common condition,
patient’s condition can be treated effectively with stimulation thera- such as failed back surgery syndrome (FBSS), could potentially allow
py or IT therapy, then neurostimulation may be considered first, sec- for comparison of the efficacy of SCS with IT therapy, but no direct,
ondary to safety considerations (42). However, there was significant controlled studies have been conducted in the past decade.
discussion among the authors surrounding the sustainability of both An algorithmic hierarchy of IT therapy for cancer pain is similar.
IT and neurostimulation therapies. Traditional spinal cord stimula- Prognosis, likely progression of the disease into different anatomic
tion (SCS) is an effective therapy, as clearly demonstrated in multiple regions/tumor characteristics, and periprocedural imaging findings
studies (44,45). However, there is a significant patient population are useful considerations for device selection. In patients with
(nearly 30%) that becomes tolerant to the therapeutic perceived cancer-related pain, Smith et al. demonstrated improvement in side-
paresthesia (46). Furthermore, Hayek and Veizi reported an explant effects and pain with IT therapy compared to conservative medical
rate from a cohort of university hospitals of nearly 23.9% (47). New management (11). Staats, in a comparative trial for AIDS and cancer
stimulation therapies may help with salvage (48–50), although there patients, showed improvement in the cancer population with IT
is a compelling argument for a role of IT therapy, certainly after SCS ziconotide (33). Furthermore, titratability and coverage/efficacy of
failure, but also before. A significant reason for SCS explant is loss of mechanical, nociceptive pain is less likely with SCS therapy (53).
therapeutic coverage or development of new areas of pain since Although coverage can be extended with traditional SCS therapy by
implant. A registry study suggested approximately 8% of patients reprogramming, repositioning of electrodes, or adding new electro-
(13/156) developed pain outside the ability of coverage of traditional des, it is not accomplished to the degree or ease of IT therapy. To
SCS at 12 months (51). Recently, data were presented at the North that end, device selection algorithmic care is suggested in Figure 3.
American Neuromodulation Society (NANS) meeting (Las Vegas, End-of-life pain device selection is typically not performed if the
2015) that suggested durability of care for >6 years with IT therapy patient has less than three months of expected longevity. The PACC

Table 5. Recommendations for Application of Intrathecal Therapy vs. Neurostimulation by the NACC Using USPSTF Criteria.

Statement Evidence level Recommendation grade Consensus level

Intrathecal therapy should be considered within the same line as III C Moderate
neurostimulation strategies to treat noncancer-related pain.
Intrathecal therapy should be considered after neurostimulation strategies III I Strong
to treat noncancer-related pain if the pain is isolated and unlikely to
spread.
Intrathecal therapy should be considered before neurostimulation therapy III C Strong
102

for active cancer-related pain that is mechanical and likely to spread.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

physical examination and a complete psychosocial evaluation


Table 6. Disease Indications for Intrathecal Drug Delivery.
(which may be optional for cancer pain) before undertaking a trial or
implant. The PACC would point out that, in the face of psychological
 Axial neck or back pain; not a surgical candidate
distress from end-of-life, not having psychological help may amplify
䊊 Multiple compression fractures

䊊 Discogenic pain
the pain experience and compound suffering.
䊊 Spinal stenosis
The cancer pain population deserves special mention, as imple-
䊊 Diffuse multiple-level spondylosis
mentation of IT therapy and medication selection, along with the
 Failed back surgery syndrome sustainability of a regimen, are largely dependent on the stage of
 Abdominal/pelvic pain the disease and life expectancy (Table 7). Deer et al. have classified
䊊 Visceral cancer patients based on categories of their disease (56). The com-
䊊 Somatic plex interplay between disease indications and patient selection will
 Extremity pain be discussed in the following section.
䊊 Radicular pain

䊊 Joint pain

 Complex regional pain syndrome (CRPS) PATIENT-SELECTION CONSIDERATIONS


 Trunk pain
䊊 Postherpetic neuralgia Updating the PACC documents of 2012 and mindful of patient
䊊 Post-thoracotomy syndromes selection, managing patient comorbidities has been a concern since
 Cancer pain, direct invasion and chemotherapy related the IDD mortality data were reported in 2009 (41). Numerous reports
 Analgesic efficacy with systemic opioid delivery complicated by have suggested best practice and careful consideration for the com-
intolerable side effects plex interplay between disease, patient characteristics, and drugs
chosen for IT delivery. As outlined previously, consideration for all
the variables and vigilance is required, and we will address them
recommends that in the future a more objective measure, such as individually (8,9,26,27,56,60–63). Some influences of determining IT
the Karnofsky Palliative Performance Scale (54) or Eastern Coopera- therapy are universal, spanning all patients considered, while others
tive Oncology Group Performance Status (55) scores be used. The require risk stratification and more granularity (Fig. 4).
medication regimen (or the development of combination therapy
strategies) may be accelerated in end-of-life pain coverage. This will
Procedural and Surgical Comorbid Disease Management
be discussed with more granularity in the medication-selection Careful consideration for comorbidities that impact wound heal-
section. ing (64) and the implementation of the therapy is crucial. Consider-
Table 5 presents PACC recommendations for application of IT vs.
ations also include ability to undergo the procedure, including
SCS therapy.
anesthesia and assessment of bleeding risk. The 2014 NACC also
reported on mitigation of surgical site infection and bleeding, with
DISEASE-SPECIFIC INDICATIONS AND an update in review at the time of this writing (65). The PACC recom-
mendations for avoiding surgical site infections appear in Table
CONSIDERATIONS 8 and those for anticoagulation management appear in Table 9.
Disease-specific indications for IT therapy have been defined pre- Implementation of IT infusion procedurally is just one aspect of
viously (8,27,56). A conceptual marriage of many factors contribute the therapy. Decision-making regarding medication, site of delivery,
to the implementation of the therapy once disease-specific indica- and infusion strategy must also be discussed. Cardiopulmonary
tions have been fulfilled, including: survival time, opioid exposure/ assessment is vital when determining the medication employed and
sensitivity, location of pain, type of pain, medication physiochemical the required vigilance surrounding the implant procedure (10,27).
properties, catheter location, pump infusion strategy, and psycho- One of the important components of IT therapy is the fear of respira-
logical features and social support of the patient. Simply stated, IT tory depression (27). It is important to understand the patient’s base-
therapy is indicated by the Food and Drug Administration (FDA) for line cardiopulmonary status, along with baseline medications that
moderate to severe trunk and limb pain, and intractable pain, where may play a role in sedation and influence the CO2 response curve by
more conservative therapies have failed (57,58). This includes a varie- shifting it down and to the right (80,81). When considering the rela-
ty of disorders, including those highlighted in Table 6. There is tive risks of oral opioids with more than 16,000 deaths per year (39),
renewed interest to cover focal extremity pain with IT therapy, but the risks of IT agents appear to be considerably less, but no compar-
although anecdotal reports exist, literature support is lacking. ative risk studies have been performed. With that said, it should be
As outlined in previous versions of the PACC and other consensus emphasized that most IT infusion patients are already opiate toler-
papers, it is imperative to have a clear diagnosis, an appropriate ant, therefore, the risk of respiratory depression in such opiate-
tolerant patients is extremely low.
Disease comorbidities that may impact the influence of opioid-
Table 7. Cancer Patient Classifications. based medications on respiratory drive include central or obstructive
sleep apnea, advanced age, pulmonary disease (obstructive or
Category 1 Category 2 Category 3 restrictive), and cardiac disease (ischemic, congestive, or myopathy
related). Obstructive sleep apnea has been observed in nearly 18
Patient with imminent Patient whose disease Patient with cancer in
million people in the United States, with physiologic responses of
death or life is stable or slowed, partial remission or
expectancy relatively with high likelihood cured, with residual
hypercapnia and pulmonary hypertension. Numerous studies sug-
short, with palliation of recurrence or chronic pain gest opioids negatively impact respiratory drive and may lead to
increased apnea duration (82–85). Age plays a role in the sensitivity
103

primary objective progression


to the depressive effects of opioids (85). The American Society of

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

Figure 4. Algorithm of patient-selection characteristics. Green arrows indicate affirmation or positive response, red arrows signify negative response. Dashed
arrows signify similarity amongst groups. Cancer pain is tiered to three categories (see Table 7).

Anesthesiologists developed consensus guidelines to mitigate risk reported surrounding delayed respiratory depression with acute IT
of opioid-related respiratory depression in the acute pain popula- opiate delivery are different from those with chronic infusion. Chron-
tion, and although the guidelines do not translate to the chronic- ic infusion is associated with slow, low volume, and low kinetic ener-
pain, opiate-tolerant patient per se, the theme is vigilance (84). Risk gy delivery wherein local rostrocaudal IT distribution is limited.
stratification of patients when employing IT therapy is imperative to Redistribution of CNS-active, IT-delivered drugs out of the IT space is
provide safe pharmacologic care. likely the culprit for respiratory effects (80,81). This concept is corrob-
Given our current knowledge, understanding the role that the orated by various recent works (8,27,55).
concomitant use of central nervous system (CNS)-active medications This new understanding has been validated in animal models,
has on IT therapy is crucial. Careful appreciation for special popula- with evidence suggesting the same mechanism in humans (93). Sim-
tions of patients is also needed, including those with kidney or liver ilarly, previous exposure is relevant. More patients are taking system-
disorders, along with the geriatric population, as clearance and elim- ic opioid medications than ever before in the United States, with
ination (pharmacokinetics) of these CNS-active medications should increased mortality. Diversion and misuse contribute to the associat-
be considered. It is beyond the scope of this text to describe the ed morbidity and mortality (40,94–96). The California State Medical
pharmacokinetics of systemic medications, but it is important to Board has created guidelines for systemic opioids to be less than 80
note that potential interaction exists between IT-delivered medica- morphine equivalents (MEs), while others advocate for less than 100
tions and CNS-active medications. Any CNS depressant can augment or 120 MEs (95,97). The CDC recently recommended chronic mainte-
opioid-induced respiratory depression (Table 10). Creatinine kinase nance doses of systemic opioids of 50 ME or less, with extra vigi-
(CK) levels need to be checked at baseline and even intermittently lance and risk assessment at >90 ME. The history of systemic
during therapy with initiation of ziconotide, as clinical trials showed exposure is important when considering IT therapy, not just for cal-
that levels can rise to two or three times the upper limit of normal. culating the total amount of systemic opioids, but also for consider-
Causation and clinical significance of this rise is unclear, but good ing the impact of IT opioid dose escalation, as failure of systemic
clinical judgment is important and symptoms need to be carefully opioids where side effects are not an excluding factor suggests fail-
evaluated if symptomatic CK elevation exists, with reduction or ces- ure of opioids intrathecally. Preclinical work has shown cross-
sation of therapy to be considered. Registry data recently suggested tolerance between IT and systemic morphine (96).
improved tolerability of ziconotide when it was not infused with sys- In one IT opioid study of patients with chronic pain, during the
temic medications having a similar mechanism of action and func- washout period before randomization to another IT drug, the major-
tion (86). This knowledge allows for risk stratification of dosing and ity of patients were able to wean IT opioid medications but with
vigilance surrounding IT implementation. Paradoxically, nonopioid- average systemic supplementation of near 300 MEs per day (32).
na€ıve patients may have an increased margin of safety when IT ther- The patients had visual analogue scale of pain intensity (VASPI)
apy is initiated, although the relationship is nonlinear and overdose scores of >80 mm, suggesting failing IT therapy. If patients are
remains a possibility (27). maintained on large amounts of systemic opioids before starting IT
therapy, the likelihood of monotherapy failure seems to be higher
Previous Systemic Opioid Use (12,14,15,38,99,100). This concept supports the approach of weaning
Discussion of patient selection requires considering all of the or reducing systemic opioids prior to initiating IT therapy.
aforementioned factors of concurrent medications, disease states, This potency shift is not predictable based on previous models for
prior treatments, and device implementation. Notwithstanding, the systemic to IT conversion (27), and there are reports of tolerance
concept of bulk flow within the IT space is limited (8,20,90–92), with reversal in as little as a week off IT agents (42). Veizi et al. reported a
potentially very little spread from the distal end of the catheter with reduction of dose escalation of nearly three times when employing
104

the CSF. It has long been appreciated that the supratentorial effects a local anesthetic (99). Many authors recommend weaning strategies

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Table 8. Recommendations for the Avoidance of Surgical Site Infections as Recommended by the PACC.

Recommended practice Origin of recommended References and


practice§ comment
Preoperative practices

Utilization of preoperative antibiotics for trials CDC IA and NICE Bowater et al. (66) demonstrated that antibiotic
prophylaxis is effective for reducing the risk of wound
infection for all types of surgery.
Utilization of preoperative weight-based antibiotic dosing for CDC IA and NICE Weight-based dosing of antibiotics is required to achieve
trials therapeutically effective drug concentrations (67,68).
Utilization of preoperative antibiotics for implants CDC IA and NICE Bowater et al. (66) demonstrated that antibiotic
prophylaxis is effective for reducing the risk of wound
infection for all types of surgery.
Utilization of preoperative weight-based antibiotic dosing for CDC IA Weight-based dosing of antibiotics is required to achieve
implants therapeutically effective drug concentrations (66,68).
Utilization of preoperative nasal culture and application of CDC 1A Universal decolonization seems associated with a low risk
mupirocin to prevent Staphylococcus aureus surgical site of mupirocin resistance in Staphylococcus aureus (69).
infections Mupirocin and antiseptic body wash may reduce
superficial but not deep surgical site infections (70).
Mupirocin decolonization is effective in a high-risk popula-
tion (71), however, compliance is low and resistance
occurs (72,73). Nasal povidone-iodine may be considered
as an alternative (74).
Appropriate preoperative timing (within 1 hour prior to CDC IA, NICE, SCIP
surgical incision excluding vancomycin) of prophylactic
antimicrobial administration for implants
Hair removal (when required) with electric clippers CDC IA and NICE
immediately before the surgical procedure

Intraoperative practices
Utilization of double gloving CDC II and NICE Tanner and Parkinson (75) concluded that the addition of
a second pair of surgical gloves reduces perforations to
the innermost gloves. Although there is insufficient
evidence that double gloving reduces the risk of SSIs,
NICE recommends wearing two pairs of sterile gloves
when there is a high risk of glove perforation and the
consequences of contamination may be serious (76).
Utilization of chlorhexidine gluconate for preoperative skin CDC IB and NICE CDC and NICE recommend the use of an appropriate
antiseptic agent antiseptic agent (povidone–iodine or chlorhexidine-
containing products). Darouiche et al. (77) demonstrat-
ed that preoperative skin preparation with
chlorhexidine-alcohol is superior to povidone–iodine for
preventing SSI.

Postoperative practices
Application of an occlusive dressing following a trial CDC IB and NICE CDC recommends applying a sterile dressing for 24–48
hours postoperatively (category IB). NICE recommends
interactive dressings. Hutchison and McGuckin
demonstrated lower rates of infection with occlusive
dressings (78).
Understanding maximum time criterion for defining a deep CDC Infection occurs within one year if an implant is in place
surgical site infection of an implantable device (one year) and the infection appears related to the operation.
No continuation of antibiotics into the postoperative period SCIP SCIP recommends the discontinuation of antibiotics
for trials beyond 24 hours* within 24 hours after surgery.
No continuation of antibiotics into the postoperative period SCIP SCIP recommends the discontinuation of antibiotics
for implants beyond 24 hours† within 24 hours after surgery.
CDC, centers for disease control; NICE, National Institute for Health and Care Excellence; SCIP, Surgical Care Improvement Project of the Joint
Commission; SSIs, surgical site infections.
*Examination of survey questions 22 and 23.

Examination of survey questions 24 and 25.
§
CDC recommendations. IA: Strongly recommended for implementation and supported by well-designed experimental, clinical, or epidemiological stud-
ies. IB: Strongly recommended for implementation and supported by some experimental, clinical, or epidemiological studies and strong theoretical ratio-
nale. II: Suggested for implementation and supported by suggestive clinical or epidemiological studies or theoretical rationale.
105

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

Table 9. Anticoagulation Management Practices for Intrathecal Therapy as Recommended by the PACC.

Anticoagulant Recommendation for trial Recommendation for permanent implant

Warfarin Discontinue five to seven days before, INR <1.5; if Discontinue five to seven days before, INR
bridging required, refer to bridging medication; <1.5; if bridging required, refer to bridging
continue cessation during duration of trial, medication; resume 24 hours postoperatively.
resume 24 hours following trial catheter removal.
Enoxaparin (LMWH) Hold therapeutic dose of LMWH 24 hours before Hold therapeutic dose of LMWH 24 hours
procedure; hold for duration of trial; resume 24 before procedure; resume 24 hours
hours following catheter removal. following surgery
Clopidogrel (ADP receptor antagonists) High-risk patients for cardiac events—discontinue High-risk patients for cardiac events-discontinue
at least five days before; low risk seven to ten at least five days before; low risk seven to
days before; hold for duration of trial; resume ten days before; resume 24 hours following
24 hours following catheter removal. surgery.
Prasugrel (ADP receptor antagonist) Discontinue seven to ten days prior to procedure, Discontinue seven to ten days prior to
hold for duration of trial, resume 24 hours procedure, hold for duration of trial,
following catheter removal. resume 24 hours following lead removal.
Ticlopidine (ADP receptor antagonists) Discontinue 14 days prior to procedure, hold for Discontinue 14 days prior to procedure;
duration of trial, resume 24 hours following resume 24 hours following surgery.
catheter removal.
Abciximab, eptifibatide, tirofiban Discontinue for three days prior to procedure, Discontinue for three days prior to procedure,
(platelet GPIIb/IIIa receptors) hold for duration of trial, restart 24 hours hold for duration of trial, restart 24 hours
following catheter removal.‡ following the surgery.‡
Dipyridamole, aggrenox (aspirin/ Discontinue seven days prior to procedure, Discontinue for seven days prior to procedure,
dipyridamole) (phosphodiesterase hold for duration of trial, restart 24 hours hold for duration of trial, restart 24 hours
inhibitors) following catheter removal.§ following the surgery.§
Naproxen, ketorolac, ibuprofen, Discontinue seven days prior to procedure, Discontinue seven days prior to procedure,
etodolac, etc. (nonsteroidal hold for duration of trial, reinitiate 24 hours hold for duration of trial, reinitiate 24 hours
anti-inflammatory drugs)§ following catheter removal. following the surgery
Aspirin§ Discontinue seven days prior to procedure, Discontinue seven days prior to procedure,
hold for duration of trial, reinitiate 24 hours hold for duration of trial, reinitiate 24 hours
following catheter removal. following surgery.
Herbals (ginseng, ginko, garlic) Discontinue seven days prior to the procedure, Discontinue seven days prior to the procedure;
hold for duration of trial, reinitiate 24 hours reinitiate 24 hours following surgery.
following catheter removal.
Dabigatran etexilate mesylate, Discontinue five days prior to procedure, hold Discontinue five days prior to procedure,
rivaroxaban (direct thrombin for duration of trial, reinitiate 24 hours hold for duration of trial, reinitiate 24 hours
inhibitors) following catheter removal. following surgery.
Heparin IV* On a case-by-case basis On a case-by-case basis
Heparin SQ† On a case-by-case basis On a case-by-case basis
INR, international normalized ratio; ADP, adenosine diphosphate; LMWH, low molecular weight heparin; NA, not available; IV, intravenous; SQ,
subcutaneous.
*Requires inpatient hospitalization and monitoring, suggesting a special need or indication for neurostimulation and should be assessed on case-by-case
basis.

Peaks at 2–4 hours after administration, typically thrombotic prophylaxis as inpatient and may require platelet assessment if more than four-day dosing.
Please refer to American Society for Regional Anesthesia guidelines and determine on a case- by-case basis.

Typically contraindicated four weeks following surgery. If reinitiated, careful follow-up and vigilance is suggested (79).
§
Current recommendations (79) suggest variable stoppage is necessary based on clinical context, and on the specific half-life of the nonsteroidal anti-
inflammatory in question. The half-life determines the time required for discontinuation in order to limit the drug’s effect on platelet function.

(101), with the core concept of improving patient selection by mov- requirement is somewhat obscured with the recently described
ing away from salvaging high-dose systemic opioid failure. The pre- weaning strategies, but the theme is suggested (13).
dictive value of systemic dose requirement to IT opioid dose The concept of limiting systemic opioid exposure to less than 100
MEs has already been proposed (1,38), with the CDC recommending
less than 50 ME for noncancer related chronic pain and increased
Table 10. Central Nervous System-Active Medication Classes That May vigilance above 90 MEs (102). Although no specific data comparing
Interact With Intrathecal Opioids (86–88). prior systemic dose to IT dose exist, enough prospective and retro-
spective data suggest it may be beneficial as a tool for IT dose esca-
Benzodiazepines
lation prediction (13,35). Furthermore, there does not seem to be a
Antidepressants
gender difference with opioid dose, although in one study women
Anticonvulsants
Muscle relaxants were more likely to continue to receive oral opioid adjuvants (97).
Hatheway et al. described the use of patient-controlled boluses to
106

EtOH consumption
minimize systemic opioid use with IT therapy (103).

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Table 11. Recommendations for Patient Selection Criteria for Intrathecal Therapy by the PACC Using USPSTF Criteria.

Statement Evidence level Recommendation grade Consensus level

Patients with comorbidities that negatively affect cardiopulmonary function III C High
need increased vigilance when instituting intrathecal opioid therapy.
Localized pain can be adequately covered with intrathecal therapy. II B Strong
Diffuse pain can be adequately treated with intrathecal therapy. III C Moderate
Global pain can be adequately treated with intrathecal therapy. III D Moderate
Intrathecal therapy should not be used as salvage therapy for failing II B Moderate
systemic opioids.*
*Patients who are weaned prior to the trial have a higher likelihood of sustained success (15,101). Different titration schedules have been recommended.

Sustainability of Intrathecal Opioid Therapy precise abdominal pain or axial back pain from vertebral compres-
Dose escalation with chronic IT opiates has been a cause for con- sion fracture. Diffuse pain refers to a whole extremity involvement,
cern. An early retrospective study noted that in cancer patients back or leg discomfort, abdominal pain encompassing more than
receiving IT infusion of morphine, for periods in excess of three one quadrant, and so on. Global pain is total body pain.
months after initial stabilization (4.8 mg/d), 48% showed less than a The PACC recommendations for patient selection appear in
twofold increase in dose by three months (104). In 52 cancer Table 11.
patients, initial dose (3.8 mg/d) rose by a factor of 2.5 at four months Consensus Point 6. Patients with comorbidities that negatively
(105). In 50 cancer patients, receiving infusion for a mean of 142 affect cardiopulmonary function need increased vigilance when
days (7–584 days), the mean IT morphine starting dose, 2.5 mg/day, instituting intrathecal opioid therapy.
increased to a mean final dose of 9 mg/day (106). In a series of
chronic noncancer pain patients (n 5 88), mean IT morphine dose Consensus Point 7. Localized, diffuse and global pain can be ade-
increased from 10 mg/day at 6 months to 15 mg/day at 36 months quately treated with intrathecal therapy. The evidence for global pain
after initiation of therapy (107). In another study, presence of neuro- treatment is less well defined and should be approached cautiously.
pathic pain seemed to be strongly predictive of IT dose escalation
(108). Seemingly, patients less than 50 years old have a greater Consensus Point 8. Intrathecal therapies should be used at an
chance of opioid dose escalation intrathecally compared to patients appropriate time in the algorithm and not as a salvage treatment.
older than age 50 (12). Mitigating strategies include proper patient
selection, minimizing pre-IT therapy opioid dose, proper localization
of pain, and congruent catheter placement. Local anesthetics, such
MEDICATION-SELECTION RECOMMENDATIONS
as bupivacaine have been shown to have synergistic interaction AND CONSIDERATIONS
with opiates (109,110). Veizi et al. reported dose escalation mitiga-
tion with concurrent use of bupivacaine, and although this is off- Since the publication of the PACC reports of 2012, more energy by
label, it appears logical and should be considered (99). these authors has focused on patient selection, procedure standardi-
zation, and infusion therapies compared to new medications. The
Psychological Assessment and Social Support Evaluation FDA has approved ziconotide and morphine for IT infusion for the
Previous consensus guidelines, best practices, and previous PACC treatment of pain. Hydromorphone from Mallinckrodt plc. is under-
iterations have all commented on psychological assessment of going clinical trial for potential IT labeling. Notwithstanding, the
patients for IT therapy (8,21,22,27). There has been little update on PACC of 2012 provided a framework to determine which IT medica-
patient selection surrounding this topic. Category 1 cancer patients tions to use when differentiating the patient’s pain as either nocicep-
do not need a robust psychological screen, as palliation is the goal tive or neuropathic or mixed pain. This framework of understanding
of IT therapy, however, they may benefit from counseling regarding has been expanded in 2016. Off-label monotherapy or combination
death, dying and chronic illness. For all others, clear descriptions of therapy should be considered after failure of FDA-approved medica-
tions or when these medications are contraindicated. The question
developing a partnership and setting expectations between patient
of which medication or combination of medications to use is impor-
and treatment team is suggested in the literature. Importantly, zico-
tant and is based on safety. The current PACC algorithms were creat-
notide is contraindicated in patients with a history of psychosis, and
ed to help guide clinicians in the safe and effective use of IT therapy;
alternative IT medications should be used (111).
however, physicians should use their own best clinical judgment in
making treatment decisions for their patients.
Pain Characteristics: Regional vs. Diffuse vs. Global
There is little doubt that CSF flow dynamics and the pharmacoki-
Consensus Point 9. Off-label drug monotherapy or combination
netic profiles of IT agents are better understood now than ever
therapy is not recommended until FDA-approved drugs are tried
before. Little bulk flow exists (90–92). Rostral spread within the IT
and failed or are contraindicated. In cancer pain, the on-label drugs
space is limited from the catheter tip. Although there is limited evi-
can be used during the trial phase. If the results are not acceptable
dence to support catheter placement congruent with the dermato-
due to lack of efficacy or side effects, an admixture with bupivacaine
mal area of pain, consensus remains high that this is a crucial
or the primary use of fentanyl is supported by our consensus.
component to sustained IT care. Similarly, placement of the catheter
dorsal to the spinal cord delivers medication closer to the dorsal ele- Consensus Point 10. The PACC algorithms are based on improv-
ments of the spinal cord. Localized pain examples include the band- ing safety and efficacy in clinical practice, which includes the use of
107

like dermatomal challenges of postherpetic neuralgia or unilateral off-label drugs.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

Table 12. Cancer or Other Terminal Condition-Related Pain With Localized Nociceptive or Neuropathic Pain.

Line 1A Ziconotide Morphine


Line 1B Fentanyl Morphine or fentanyl 1
bupivacaine
Line 2 Hydromorphone Hydromorphone 1 Hydromorphone or Morphine or
bupivacaine fentanyl or morphine 1 hydromorphone or
clonidine fentanyl 1 ziconotide
Line 3 Hydromorphone or Ziconotide 1 Ziconotide 1 clonidine Hydromorphone or Sufentanil
morphine or fentanyl 1 bupivacaine morphine or fentanyl 1
bupivacaine 1 clonidine bupivacaine 1 ziconotide
Line 4 Sufentanil 1 ziconotide Sufentanil 1 Baclofen Sufentanil 1 clonidine Bupivacaine 1 Bupivacaine 1
bupivacaine clonidine 1 clonidine
ziconotide
Line 5 Sufentanil 1 bupivacaine 1 clonidine
Line 6 Opioid* 1 bupivacaine 1 clonidine 1 adjuvants†
*Opioid (all known intrathecal opioids).

Adjuvants include midazolam, ketamine, octreotide.

Consensus Point 11. The algorithms are based on evidence and medications and is greatly dependent on catheter location. Neuro-
consensus on safety. The patient’s physician and good clinical judg- pathic pain generally responds to ziconotide, opioid plus local anes-
ment should guide individual patient care. thetic, local anesthetic alone, clonidine plus opioid, and clonidine
Considerations of the highlighted variables include patient diag- alone. Nociceptive pain generally responds to opioid, ziconotide,
noses and expected patient survival time (11), sustainability of the IT opioid plus local anesthetic, and local anesthetic alone. Manipulative
regimen (12,13), previous exposure to opioids (14,15), location of variables that can increase drug spread include the physiochemical
pain (diffuse vs. localized vs. global), type of pain (nociceptive, neu- properties of the drug, kinetics of the injectate, and the volume
ropathic, or mixed), the physiochemical properties of lipid solubility delivered (16,90). Increased concentration and daily doses of opioids
of the IT drugs (16,17), CSF dynamics and pharmacokinetics (18–20), (except fentanyl) are associated with granuloma formation, and care
catheter location (20), pump and catheter characteristics, kinetics of should be taken when dose escalation is rapid or doses and concen-
the IT infusate (20), and psychological status (21–23) of the patient trations are known to be reaching levels associated with granuloma
with chronic pain. formation (8,10). The possibility of granuloma formation should be
considered when employing opioid-based medications.
The PACC 2016 recommendations allow integration of the
INTRATHECAL THERAPY IN NEUROPATHIC applied 2012 PACC nociceptive and neuropathic localized and dif-
fuse pain recommendations for cancer or terminal illness
AND NOCICEPTIVE PAIN STATES (Tables 12–15), and for noncancer pain (Tables 16–19). For medica-
tion selection within the tiered recommendations, it is important to
Employing the familiar nociceptive and neuropathic pain classifi-
consider age, type of pain, and anticipated duration of therapy. In
cation for medication selection as a framework, the reorganization
cancer pain, the evidence would suggest combination therapy
of medication selection is based on many factors, including survival
might be warranted as a first- line strategy, which is a different rec-
expectation, age, previous exposure to opioids, location of pain,
ommendation from that for treatment of noncancer pain. If the
type of pain, and catheter location. The pharmacokinetics of the IT
patient responds to morphine or ziconotide as single medications
medications employed (112) point toward a potentially greater
during a trial, we would still recommend that on-label drugs be
spread with multiple bolus delivery compared to continuous
used initially. This distinction is highlighted by the evidence-
infusion. weighted tier system designation of line 1A and 1B. Line 1A repre-
Nociceptive and neuropathic pain, regardless of age, expected sents medication with level I evidence. Furthermore, these medica-
survival, or pain location, may respond to a combination of tions are FDA approved, with the intent of honoring the evidence
available. However, if the patient does not respond to on-label
Table 13. Cancer or Other Terminal Condition-Related Pain With
monotherapy during the trial phase, then fentanyl and combination
Localized Pain: Evidence Level, Recommendation Strength, and Consen- therapies, including admixtures with bupivacaine, are supported by
sus Level. the consensus. Baclofen is FDA approved for spasticity and is some-
times helpful in managing pain associated with spasticity.
Tier Evidence level Recommendation grade Consensus level Titratability for patients with cancer pain (Category 1 and 2, see
Table 7) is extremely important. There is significant evidence that
Line 1A I A Strong suggests opioid 6 bupivacaine is helpful in this population
Line 1B II-1 B Strong
(113,114). Careful attention should be made by caregivers to escala-
Line 2 II-3 B Strong
tion of dose or concentration above certain recommended levels (8).
Line 3 III C Moderate
Line 4 III I Weak
The tiered selection of the recommended medications was based
Line 5 III I Weak on levels of evidence surrounding the safety and efficacy of the med-
ication provided. Ziconotide monotherapy and opioid 1 bupivacaine
108

Line 6 III I Weak


share level I evidence for their use in the cancer population

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Table 14. Cancer or Other Terminal Condition-Related Pain With Diffuse Nociceptive or Neuropathic Pain.

Line 1A Ziconotide Morphine


Line 1B Hydromorphone Morphine or hydromorphone 1 bupivacaine
Line 2 Hydromorphone or Morphine or hydromorphone 1 ziconotide
morphine 1 clonidine
Line 3 Hydromorphone or Ziconotide 1 bupivacaine Ziconotide 1 Hydromorphone or Sufentanil
morphine or fentanyl 1 clonidine morphine or fentanyl 1
bupivacaine 1 clonidine bupivacaine 1 ziconotide
Line 4 Sufentanil 1 ziconotide Baclofen Sufentanil 1 Sufentanil 1 clonidine Bupivacaine 1 clonidine 1 Bupivacaine 1 clonidine
bupivacaine ziconotide
Line 5 Sufentanil 1 bupivacaine 1 Sufentanil 1 Sufentanil 1 clonidine 1
clonidine bupivacaine 1 ziconotide
ziconotide
Line 6 Opioid* 1 bupivacaine 1 clonidine 1 adjuvants†
*Opioid (all known intrathecal opioids).

Adjuvants include midazolam, ketamine, octreotide.

(11,33). Little evidence suggests that ziconotide is helpful in com- Consensus Point 14. Ziconotide has strong clinical evidence for
bination with opioids to manage cancer pain. No prospective, con- efficacy.
trolled studies of ziconotide plus opioids have been performed.
Consensus Point 15. There are no cases of death from ziconotide
Stability of admixtures becomes a concern when multiple medica-
overdose and no granuloma formation has been reported.
tions are used for combination therapy (115). There is also concern
that drug mixing may increase permeability of medications into Consensus Point 16. Unless contraindicated, ziconotide should
the pump rotor, resulting in corrosion and pump failure (58). be the first drug selected in the population of noncancer patients
Reports of ziconotide plus bupivacaine have little supportive evi- discussed in this consensus.
dence. For palliative reasons in the cancer population more com-
binations of medicines are trialed, and Tier 6 was added to the
PACC 2012 algorithm. When contemplating higher-tiered recom-
RECOMMENDED STARTING DOSAGES
mendations, it is crucial to consider the category of cancer pain
Starting dosage ranges of IT medications recommended by the
that the patient has (see Table 7). PACC panel have not changed since the PACC of 2012 (Tables 20
Consensus Point 12. The disease process should be considered and 21). These doses assume chronic continuous infusion. Bolus
when making decisions on algorithms for patient care. strategies have been reported (1,116), but there are limited data to
support widespread adoption. IT dosing studies with bolus-only or
Consensus Point 13. The stage of cancer and survival time bolus-weighted infusion strategies are presently ongoing (35).
should be considered when considering drug titration. Appropriate starting opioid dosages may vary according to the
Recommendations for medication selection for noncancer local- patient’s baseline oral intake at the time IT therapy is initiated, and it
ized pain need to be approached mindfully, and age and pain type is suggested that patients be stratified by risk regarding cardiopul-
should be carefully considered. As can be seen in the noncancer- monary depression and site of service initiation. Conservative initia-
pain tiered algorithm, and assuming that the catheter location is tion dosing strategies are recommended.
congruent to the painful area, medication recommendations are It is important to consider morbidity and mortality data of IT deliv-
based on the physiochemical properties of the drug. Built into the ery when gauging appropriate starting doses. Coffey et al. reported,
algorithms are pathways for diffuse or local, nociceptive, and neuro- from device registration and Social Security death master file analy-
pathic pain types. The evidence behind Tiers 2–5 (or 6 for cancer or ses, an IT opioid therapy mortality rate of 0.088% at three days after
end-of-life pain) is largely dependent on experience/consensus from implantation, 0.39% at one month, and 3.89% at one year (41). All
the PACC members, with graded strength of consensus. patients were initiated on an opioid dose of >0.5 mg/day. It is also
important to contrast IT drug delivery mortality with that for pre-
scription drug overdose death rates, which quadrupled between
Table 15. Cancer or Other Terminal Condition-Related Pain With
2000 and 2014, from 1.5 to 5.9 deaths per 100,000 people (117).
Diffuse Pain: Evidence Level, Recommendation Strength, and Consensus
Level. Site of service for trialing and dosing of IT therapy is an important
issue when considering morbidity and mortality. This article is
Tier Evidence level Recommendation grade Consensus level accompanied by a PACC recommendation article that pays exclusive
attention to trialing (118), where similar clarity is required. Conserva-
Line 1A I B Strong tive dosing, regardless of patient risk assessment, is highly recom-
Line 1B II B Moderate
mended in both articles. Risk assessment not only includes the
Line 2 II-3 B Strong
biologic disease indications and patient-selection criteria aforemen-
Line 3 III C Moderate
Line 4 III I Weak
tioned, but also the nonbiologic site of service where trialing and
Line 5 III I Weak dosing changes occur. It was recommended in the previous PACC to
perform opioid trials and initiate monotherapy or combination opi-
109

Line 6 III I Weak


oid therapy followed by a 23-hour observation period, which

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

Table 16. Noncancer-Related Pain With Localized Nociceptive or Neuropathic Pain.

Line 1A Ziconotide Morphine


Line 1B Fentanyl Fentanyl 1 bupivacaine
Line 2 Fentanyl 1 clonidine Hydromorphone or morphine 1 Fentanyl 1 bupivacaine 1 clonidine Bupivacaine
bupivacaine
Line 3 Fentanyl 1 Morphine or hydromorphone 1 Ziconotide 1 clonidine or Bupivacaine 1
ziconotide 1 clonidine bupivacaine or both clonidine
bupivacaine
Line 4 Sufentanil 1 bupivacaine or clonidine Baclofen Bupivacaine 1 clonidine 1 ziconotide
Line 5 Sufentanil 1 bupivacaine 1 clonidine Sufentanil 1 ziconotide

typically requires hospital admission. In review of publications and been revised, by consensus, to six hours as long as there are no
consensus opinion since that publication, we have modified our rec- signs of neurologic dysfunction prior to initiation. The risk of mor-
ommendation so that the site of service for IT therapy be based on phine overdose applies when using higher initial drug concentra-
several mitigating factors. tions of morphine. This is important since the FDA considers drug
In the perioperative setting for trials, the safe IT dose of morphine dilution as an off-label use of drug. In settings where the initial drug
was determined in a meta-analysis to be near 0.075 and 0.15 mg for concentrations create the need for a starting dose outside of the
a single IT bolus, although the disparity of the definition of clinically PACC recommendations, an overnight admission is advised.
significant respiratory depression muddies the ability to determine No evidence suggests superiority of one trialing method over
the exact incidence (119). In a retrospective comparison study of another, which includes duration of trial (15,124). However, inpatient
parturients in labor investigating opioid-related side effects of mor- catheter trials offer the flexibility to trial different intrathecal medica-
phine or bupivacaine, the incidence of opioid-related side effects tions and regimens following one dural puncture, and may be help-
did not statistically differ (pruritus, nausea, vomiting, respiratory ful in the complex patient.
depression). None of the 114 patients in this study had respiratory Special comment needs to be made here regarding patient-
depression at doses of 0.04 mg hydromorphone or 0.1 mg morphine controlled bolus administration and IT opioid rotation or a medica-
when observed for no less than 24 hours (120). In an assessment of tion switch. The 2012 PACC recommendations suggested that
1524 postoperative patients, one patient who received a single- patient-controlled dosages be 5–20% of the total daily dosage. In a
bolus IT dose had less than ten breaths per minute (121). In a pro- retrospective review from the Cleveland Clinic, up to 30% of the
spective study comparing morphine and fentanyl in the parturient 24-hour dose could be administered safely during each patient-
population for cesarean delivery, there were no respiratory depres- activated dose, up to four times daily (125). This represents a signifi-
sion events defined as less than ten breaths per minute (122). In a cantly larger incremental increase of the 24-hour dose, suggesting
comparison of spinal analgesia for transurethral surgical procedures, that more prospective or retrospective data are required. In another
in patients who received 25 mcg of fentanyl intrathecally, no patient unpublished study from the Cleveland Clinic, patient-controlled IT
(n 5 20) experienced respiratory depression or complications (122). analgesia proved to be cost-effective, paying for the IT device in
Although not specific to the chronic pain patient undergoing IT trial eight months (126).
for candidacy of IT chronic delivery, evidence suggests low-dose opi-
Consensus Point 17. The initiating dose of intrathecal opioids
oid trialing is safe in the outpatient setting.
and ziconotide should be as low as reasonably expected to provide
Does the safety profile described during IT trialing translate to the
analgesia.
initiation of therapy as an outpatient? The mortality data for IT thera-
py includes data from implantation, revision, and refill of the device Consensus Point 18. The initiating dose of intrathecal opioids
(41). There are no data suggesting safety or danger of IT opioid initi- and ziconotide delivered continuously should be 50% or less of the
ation in an outpatient setting. However, it is suggested by expert dose used during bolus trialing.
consensus that the 24-hour initiating IT dose be half of the effica-
Consensus Point 19. The PACC recommends that the primary
cious/successful trialed IT opioid dose. For ziconotide, the previous
medication be weaned and discontinued when converting medica-
recommendation of a 12-hour observation period after initiation has
tions from one single medication to a different single medication in
the algorithm. The use of ziconotide and bupivacaine do not have
Table 17. Noncancer-Related Pain With Localized Nociceptive or Neu-
risk of withdrawal and weaning is not needed. The abrupt stopping
ropathic Pain: Evidence Level, Recommendation Strength, and Consen-
of an opioid is not recommended.
sus Level.
Consensus Point 20. The PACC recommends careful attention to
Tier Evidence level Recommendation grade Consensus level side effects when adding any adjuvant drug to a primary drug.
Line 1A I A Strong Consensus Point 21. Medications with significant withdrawal syn-
Line 1B II-3 B Strong dromes, including clonidine and baclofen, require rescue strategies
Line 2 II-3 B Strong
in the event of abrupt cessation or interruption in intrathecal
Line 3 III C Moderate
Line 4 III I Weak
delivery.
Line 5 III I Weak Maximal recommended daily doses based on preclinical studies,
animal toxicity studies and consensus were published in the 2012
110

Line 6 III I Weak


PACC. Preclinical work with fentanyl and sufentanil used single

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Table 18. Noncancer-Related Pain With Diffuse Nociceptive or Neuropathic Pain.

Line 1A Morphine Ziconotide*


Line 1B Hydromorphone Morphine or hydromorphone 1
bupivacaine
Line 3 Hydromorphone or Fentanyl 1 bupivacaine Ziconotide 1 morphine or
morphine 1 clonidine hydromorphone
Line 4 Hydromorphone or Fentanyl 1 ziconotide Sufentanil 1 bupivacaine or clonidine Ziconotide 1 clonidine or
morphine 1 bupivacaine 1 bupivacaine or both
clonidine
Line 5 Fentanyl or sufentanil 1 bupivacaine 1 clonidine Sufentanil 1 ziconotide Baclofen
Line 6 Opioid 1 ziconotide 1 bupivacaine or clonidine
*Ziconotide should be first choice in patients with >120 morphine equivalents or fast systemic dose escalation, in the absence of history of psychosis.

bolus, multiple daily boluses, or continuous infusions across many per day level safely and possibly with efficacy, however, the panel
animal models (127–133). It appears that high concentrations of fen- does recommend this dosage currently as a reevaluation milestone.
tanyl and sufentanil are tolerated, exclusive of a single injection Similarly, evidence suggests that the much higher doses of bupi-
sheep study (133). Very limited continuous infusion studies exist. vacaine are well tolerated and average concentrations are reported
Although some data suggest that 50 mcg/mL has limited data sup- in many studies (99,134,135). For all of the other drugs, the recom-
porting safety, the 2000 mcg/mL study also suggests that neurotox- mendations established in 2012 are still supported in the evidence,
icity may not be a concern, with conflicting reports with fentanyl up and we endorse the same dosing recommendations in 2016
to concentrations of 5000 mcg/mL (127,128). (Table 22).
Although these animal toxicity studies do support the safety of
Consensus Point 22. Before proceeding with aggressive dose
high concentrations of fentanyl and sufentanil, there was concern
titration above 1000 mcg per day of IT fentanyl, clinicians should
among PACC members about the phraseology of the “no known
closely monitor the outcome of each dose increase and, if efficacy is
upper limit” recommendation. Those expressed concerns included:
not being established, consider dose reduction with consideration
1) high daily doses may lead to opioid- induced hyperalgesia; and 2)
of intrathecal tolerance and hyperalgesia.
high plasma levels of the medication from very high IT doses may
limit the advantages of targeted drug delivery to the spinal dorsal
horn. The majority of PACC members agreed that in most clinical set- VARIABLES AFFECTING CHRONIC
tings patients would not benefit from daily doses higher than 1000
INTRATHECAL THERAPY
mcg of fentanyl or 500 mcg of sufentanil. Members also agreed that
the lowest possible concentration should be used, as human data Spinal Anatomy and CSF Dynamics Relevant to IT Drug
are limited. Therefore, the consensus was that a more conservative Delivery
approach to dosing of both fentanyl and sufentanil should be taken. Meninges are morphologically and physiologically implicated in
The consensus panel did note the experience of several members mechanical, immunologic, trophic, metabolic and thermal protec-
suggesting that escalation of IT fentanyl doses was often associated tion of the brain and spinal cord. In relation to spinal drug delivery,
with diminishing returns. Several have observed clinical responses the spinal meninges represent the main barrier to the transfer of
consistent with hyperalgesia and lack of clinical efficacy in dose drugs between the CSF and the spinal cord. Therefore, it is necessary
ranges above 1000 mcg per day of IT fentanyl. Further systemic to know if any of the meninges cause resistance or limitation to the
absorption of this highly lipophilic opioid may approach systemic lev- free circulation of CSF, presenting a barrier or compartmental limita-
els seen with transdermal systemic applications (>3 ng/dL) as higher tion. The spinal dural sac contains the subarachnoid space with the
IT doses are utilized. While there is no conclusive data to guide the trabecular arachnoid, the pia mater, and the subpial tissue. Drugs
panel, the PACC does suggest strong reevaluation and consideration must cross all of these structures before reaching their final target,
of other approaches as doses cross the 1000 mcg per day dosing
threshold. Clearly clinicians have utilized doses above the 1000 mcg
Table 20. Recommended Starting Dosage Ranges of Intrathecal
Medications for Long-Term Therapy Delivery.
Table 19. Noncancer-Related Pain With Diffuse Nociceptive or Neuro-
pathic Pain: Evidence Level, Recommendation Strength, and Consensus Drug Recommendation of starting dose*
Level. Morphine 0.1–0.5 mg/day
Hydromorphone 0.01–0.15 mg/day
Tier Evidence level Recommendation grade Consensus level Ziconotide 0.5–1.2 mcg/day (to 2.4 mcg/day per
Line 1A I A Strong product labeling)
Line 1B II B Strong Fentanyl 25–75 mcg/day
Line 2 II-3 B Strong Bupivacaine 0.01–4 mg/day
Line 3 III C Moderate Clonidine 20–100 mcg/day
Line 4 III I Weak Sufentanil 10–20 mcg/day
Line 5 III I Weak *Starting doses of continuous intrathecal delivery should be half of
111

Line 6 III I Weak the trial dose for opioid-based medications.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

histochemical profile, positive epithelial membrane antigen. In the


Table 21. Recommended Doses for Intrathecal Bolus Trialing.
pial layer, collagen fibers and fibroblasts continue under the pial
cells to form the subpial compartment. The trabecular arachnoid
Drug Recommended dose*
surrounds the structures inside the subarachnoid space, including
Morphine 0.1–0.5 mg the spinal cord, nerve roots and vessels that are found free within
Hydromorphone 0.025–0.1 mg the space, providing cover sheaths to these structures (144,145).
Ziconotide 1–5 mcg These sheaths are very fragile and break easily if dissected. The char-
Fentanyl 15–75 mcg acteristics of the arachnoid sheaths in the cauda equina are variable;
Bupivacaine 0.5–2.5 mg some are lax while superimposed planes of the same components
Clonidine 5–20 mcg
with a more compact appearance form others. The thickness of an
Sufentanil 5–20 mcg
arachnoid sheath ranges from 10 to 60 mm (144,145). In some cases,
*Starting doses of medication in the opioid-naive patient for outpa- a single arachnoid sheath encloses one or more nerve roots and in
tient bolus delivery do not exceed 0.15 mg morphine, 0.04 mg hydro- others the nerve root has no sheath at all (144–146).
morphone, or 25 mcg fentanyl.
It is possible that a microcatheter, with small diameter, could be
introduced inside the arachnoid sheath. By contrast, a 20G catheter,
used commonly in epidural techniques, is more difficult to intro-
the substance of the spinal cord. Dura mater, arachnoid and pia duce. If a drug is injected inadvertently or by accident inside the
mater are differentiated structures morphologically, with different sheath, the drug would have a limited dilution with CSF and, there-
properties and, therefore, must be considered and analyzed fore, could potentially be neurotoxic. Taking into account the meth-
independently. od of administering drugs, continuous injection of local anesthetic
Dural Sac through a microcatheter into these arachnoid sheaths could poten-
The dura mater is the most external layer of the dural sac and is tially be more devastating than a single injection. This is because
responsible for 90% of its total thickness. This fibrous structure, aris- repeated doses of small volumes may be accommodated inside the
ing from the meningeal fibroblast, represents a collagenous mem- sheath, leading to nerve damage. The injection of a single larger vol-
brane that confers a barrier to diffusion defined by the molecular ume instead would promote leakage of the anesthetic outside the
weight of the compound crossing the membrane (136). The remain- sheath, decreasing its potential for injury.
ing internal 10% of the dural sac is formed by the arachnoid lamina,
Lumbar Subarachnoid Ligaments
which is a cellular lamina that adds very little extra mechanical resis-
Trabecular arachnoid and subarachnoid ligaments may be related
tance to the compound movement (137). The arachnoid lamina is
to embryonic tissue remnants found in the subarachnoid space,
semipermeable, and influences the passage of lipophilic substances
where the cellular component is progressively replaced by fibrous
through the dural wall (136). The arachnoid limits the diffusion of
connective tissue. These ligaments anchor the lateral, anterior and
injected drugs to the epidural space. Dura mater has a thickness of
posterior sides of the spinal cord to the dural sac (147,148). Sub-
about 0.35 mm (0.25–0.40) (138) that it is fairly constant along the
arachnoid ligaments are similar to trabecular arachnoid, although
length of the spinal cord, with some small variations. It is comprised
contain more collagen fibers, and therefore more resistant to
of concentric dural laminas containing fibers distributed randomly in
mechanical forces. A number of 21 dentate ligaments from each
all directions (139–142). The arachnoid lamina has a thickness of 50–
side of the spinal cord hold to the dural sac. Each ligament is com-
60 microns (mm) (143). Its barrier effect is due to arachnoid cells
posed of a flat fibrous membrane between the anterior and the pos-
strongly bonded by specific membrane junctions. This cell layer rep-
terior nerve roots, and its medial edge is in direct contact with
resents a small thickness of about 10–15 mm.
subpial tissue covering the spinal cord. Laterally, these ligaments
Trabecular Arachnoid give rise to pyramidal projections that attach nonuniformly to the
The trabecular arachnoid originates from the stratum of inner cells arachnoid lamina. The most cephalic ligament is found opposite the
of the arachnoid lamina. These cells surround bundles of collagen margin of the foramen magnum between the vertebral artery and
fibers that form the axis of the arachnoid trabeculae. Near the spinal the hypoglossal nerve. The lowest dentate ligament lies between
cord, the arachnoid cells of the trabecular structure are mixed with the exit of the 12th thoracic and first lumbar spinal nerve roots; this
pial cells from the pia mater. Both types of cells share the same ligament is a thin band stretching downwards from the medullary
cone. Less commonly, posterior ligaments (posticum) are found giv-
ing shape to thin, inconsistent bands that attach the spinal cord to
Table 22. Maximum Concentrations and Daily Doses of Intrathecal the inner surface of the dural sac (147,148). There are also less resis-
Agents as Recommended by PACC 2012 (8) and 2016. tant fenestrated posterior-lateral ligaments, extending more laterally
from the dorsal roots to the arachnoid lamina. Both posterior and
Drug Maximum concentration Maximum dose per day posterior-lateral ligaments extend longitudinally from the cervical to
Morphine 20 mg/mL 15 mg the midthoracic or lumbar level. The thinner ventral ligament is
Hydromorphone 15 mg/mL 10 mg found in the anterior side of the subarachnoid space. These sub-
Fentanyl 10 mg/mL 1000 mcg arachnoid ligaments do not limit free flow of CSF in most patients,
Sufentanil 5 mg/mL 500 mcg due to their discontinuity along the dural sac.
Bupivacaine 30 mg/mL 15–20 mg*
Clonidine 1000 mcg/mL 600 mcg Pia Mater
Ziconotide 100 mcg/mL 19.2 mcg The structure of the pia mater includes a cellular layer and a sub-
pial compartment. The cellular layer is made of flat overlapping pial
*May be exceeded in end-of-life care and complicated cases as deter-
cells with a smooth and bright appearance. It is three to five pial
112

mined by medical necessity.


cells thick (10–15 mm) at medullary level and two to three cells thick

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Table 23. Cerebrospinal Fluid Volume and Nerve Root Volume (mL) per Vertebral Segment (159).

Sacral L5 L4 L3 L2 L1 T12

Cerebrospinal fluid (mean 6 SD) 2.2 6 0.6 4.8 6 1.3 5.1 6 1.1 4.9 6 0.8 5.7 6 0.9 5.8 6 1.6* 4.76 1.3*
Nerve root (mean 6 SD) 0.1 6 0.1 0.6 6 0.3 1.3 6 0.3 1.8 6 0.5 2.0 6 0.8 2.4 6 0.5* 2.4 6 0.6*
*Includes spinal cord volume.

(3–4 mm) at nerve root level. Amorphous fundamental substance is elevation of intra-abdominal pressure. The oscillatory CSF pulsations
found around pial cells. The cells measure on average 0.5–1 mm have a significant impact on the spreading of drugs after subarach-
(149,150). noid injection (156). CSF flow dynamics reveal latencies of the systol-
The subpial compartment has large amounts of collagen fibers, ic and diastolic peaks of cervical and lumbar CSF pulsations, contrary
amorphous fundamental substance, fibroblasts, and a small number to the hypothesis of a continual wave theory. Furthermore, fast flow
of macrophages, as well as blood vessels. The subpial compartment velocity reappears in the thoracolumbar spine, correlating to a large
is enclosed between the pial cellular layer and a basal membrane in respiratory influence in the thoracolumbar spine (18).
contact with neuroglial cells. The subpial compartment from low Magnetic resonance imaging (MRI) allows the estimation of CSF
thoracic vertebrae has a thickness of 130–200 mm, and here mea- volumes from human axial images under physiological and patho-
surement variations are more significant than in the pial cellular lay- logical conditions (157–160). There is a great variability of CSF vol-
er. The thickness of the pia mater is reduced to 80–100 mm at the ume between patients, although this variation also depends on the
level of the medullary cone and continues to diminish down to 50– method used to study the CSF. Sullivan et al. (159) in 2006 estimated
60 mm in the origins of the cauda equina. At nerve root level, the a CSF volume of 35.8 6 10.9 mL (range 10.6–61.3 mL) between a
thickness of the subpial compartment is 10–12 mm (149,150). perpendicular plane in the intervertebral midpoint of T12 to L1 and
At the level of the medullary cone, there are perforations or fenes- the lowest limit of the dural sac. Edsbagge et al. (160) in 2011 stud-
trations over the entire surface of the cellular layer of the pia mater. ied the complete spine and found a total CSF volume of 81 6 13 mL
These fenestrations have circular, ovoid, or elliptical shapes. While (52–103 mL). In the cervical region, there was 19 6 4 mL, in the tho-
the dimensions of these fenestrations vary, most of them measure racic region, 38 6 8 mL, and in the lumbosacral region 27 6 8 mL.
12–15 mm in length and 4–8 mm in width. At nerve root level, the Another group estimated that the total volume of CSF from L5-S1 to
pia mater also shows similar fenestrations but smaller in size (1– T11-T12 was 36.1 6 6.7 mL (161). These individual differences of CSF
4 mm) (144,149,150). volume affect the final concentration of a local anesthetic drug
Surrounding the pial cells there are numerous macrophages. The administered in the dural sac of different individuals, even with the
macrophages and other inflammatory cells seen within the pia same dose, volume and concentration given. Therefore, having con-
mater could possibly originate from subpial and subarachnoid blood sidered other relevant factors such as position of the patient or ver-
vessels, although a small proportion of them could originate from tebral level selected for subarachnoid injection, it may be that doses
immature pial cells as a result of an unknown stimulus. Probably the below 7.5 mg of bupivacaine do not ensure an adequate level of
fenestrations found in the pia mater are related to the migration of blockade in all patients.
some immature pial cells as part of an inflammatory response (151).
The number of cell junctions between pial cells is much lower Nerve Roots, CSF, and Subarachnoid Catheters
than among arachnoid cells. For this reason, pia mater is a perme- The relationship between CSF volume and nerve root at each ver-
able structure allowing the passage of drugs through intercellular tebral level is an unknown subject that may be of interest when we
spaces. However, in the area of the conus medullaris the permeabili- consider the concentration of drugs in CSF and the amount of nerve
ty could be higher if the fenestrations are present in the patient. tissue that must be crossed (161–163). In the cadaver it is possible to
measure the volume of each nerve root, but more difficult to deter-
Cerebrospinal Fluid mine the amount of CSF related to each nerve root. Recently Prats-
The volume of the CSF has obvious relevance as a determinant of Galino et al. estimated the volumes in the segments from L5-S1 to
dilution of drugs in the subarachnoid space (152). About 500 mL of T11-T12 (162). The total volume of CSF was 29.95 6 5.66 mL
CSF is formed each day, mainly by the choroid plexuses of the cerebral (Table 23) and the volume of the nerve roots was 10.38 6 2.4 mL.
ventricles with uncertain contribution from ependyma, pia, and brain The total mean volume of CSF at each lower thoracic and lumbar
parenchyma. A small proportion of CSF leaves the skull and enters the level is around 5 mL per segment, but with a wide range of results
spinal subarachnoid space, passing downwards, posterior to the spinal between the different levels (162) (Table 23).
cord and returning upwards, anterior to the spinal cord (153), with lit- The existence of concentration gradients for many compounds in
tle bulk flow. The rate of absorption through the arachnoid villi varies the CSF along the spinal canal was established in the 1990s
and is adjusted to maintain a pressure within normal range. (139,140). These gradients imply de facto that the CSF cannot be a
There are oscillations of the CSF pressure, which are synchronized circulatory system, and recently MRI evidence has confirmed that
with intracranial arterial pulsations for both respiratory and circulato- movements of the CSF through oscillations rather than a flow (139).
ry motors. These changes of pressure could help the dilution of The velocity of CSF oscillation waves has been calculated at 4.6 m/
drugs injected in the CSF to reach a homogenous concentration sec (SD 1.7 m/sec) (140). This new knowledge has been surprising to
around the nerve roots and spinal cord. Their amplitude is about many practitioners and should be considered in all future pharmaco-
9 mm per cycle in the cervical CSF and about 4 mm at the thoracic- kinetic studies of IT drugs.
lumbar junction, with minimal movement in the distal part of the Very little is known about the mechanisms of drug dispersion in
113

lumbar sac (154,155). Pulsations probably increase with the the human CSF. The best available in vivo data are still derived from

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

a sophisticated porcine model developed by Bernards (16,20,152). liquid/solid interfaces (168). Velocity and amplitude variations also
Overall these studies suggest that all drugs that have been tested occur in various locations within the spinal canal (169).
for IT infusion and are relevant to the algorithms are poorly dissipat- In a small-volume (spinal anesthetic) CFD model, speed of drug
ed in the CSF. This is due to a combination of factors including the transport (i.e., mixing) was strongly affected by the frequency and
low kinetic energy (low flow rate) provided by all implantable volume of CSF pulsations (156). Large-volume bolus injection of
pumps currently on the market, limited liquid/liquid diffusion coeffi- more than 10% of total estimated CSF volume has been shown to
cient, and the absence of meaningful CSF bulk flow. Local IT mecha- result in rapid substantial mixing throughout the entire spinal axis
nisms related to the interaction of the oscillation of the CSF across independent of pulsatile mixing, but this is orders of magnitude
obstacles, such as nerve roots and ligaments, as well as irregularities greater than clinically used in simple continuous or intermittent
in diameters and contours of the walls of the spinal canal, combine patient-activated bolus mode through an implanted catheter (164).
to form regional vortices that increase the dispersion of drugs by Many animal studies have shown a rostral-caudal gradient from
several orders of magnitude (164). the catheter tip (20,81,91), and the recent work of Wallace and Yaksh
confirms this in a human study (170). In patients receiving IT mor-
phine, CSF morphine concentrations decreased by distance from the
CSF Dynamics catheter tip with a gradient that correlated with the infusion dose,
Factors that affect the dynamics of CSF flow include heart rate, and over a range of infusion rates of 0.1–1.0 mL/day.
blood pressure (156), and the pulmonary ventilation, which appears Just as volume and flow rate of injection in spinal anesthetics
to be one of the most important drivers (165). Traditional knowl- compared to continuous catheter infusion limit the applicability of
edge and experience with local anesthetics, opioids, and novel spinal anesthetic data, such discrepancies may also explain the
agents administered in the lumbar spinal subarachnoid space for apparent initial “failures” of IT drug delivery when the catheter loca-
surgical anesthesia and analgesia unfortunately contribute little tion, volume, and rate of delivery vary from the trial methodology
understanding to the complexities of IT catheter-targeted drug and lead to varying and lesser drug distribution reflecting pharma-
delivery for chronic pain and spasticity management. In the former, cokinetic differences. This has been described with the lipophilic
technical factors such as bolus injection speed or rate, volume, baric- drug bupivacaine (171), and when simple addition of lipophilic bupi-
ity, and patient factors such as height and posture, significantly influ- vacaine to an existing hydrophilic opioid pump mixture was shown
ence drug distribution. In the latter, drug distribution is highly to be of no benefit (172).
influenced by CSF flow dynamics, where multiple recent studies Given all these variables, the amount of drug present at a particu-
have led to a new understanding of drug distribution, which in time lar site along the neuraxis distant from the injection site is difficult to
may lead to improved efficacy and safety of IT drug delivery. determine and is not likely to be uniformly distributed from a CFD
The intended action of intrathecally delivered drugs is the spinal perspective (167). Experimentally, drug distribution is limited to a
cord and to a lesser extent the brain, and they are delivered into the few centimeters around the tip of the catheter (171,173), and disper-
CSF through a catheter connected to an implanted pump. The CSF sion around the cord is also limited (90). This leads to factors to con-
is secreted by the choroid plexus and brain parenchyma at a rate of sider when placing a permanent catheter.
0.3–0.4 mL/min and the total volume ranges from 90 to 150 mL.
Approximately a third of the total volume is contained in the com- Catheter Location and Placement
pliant spinal subarachnoid space. Choroid plexus and brain paren- The traditional teaching of many instructors has reasonably rec-
chyma are not the only sites of CSF production, as glial cells, water ommended placing the catheter tip close to the target receptors of
transporters know as aquaporins, and other bidirectional mecha- the spinal segment(s) associated with the dermatome/sclerotome/
nisms produce flow rates greatly exceeding traditional net CSF viscerotome of the primary pain generator. This issue of delivery
secretion rates (166). Absorption traditionally has been considered location reflects on: 1) the fact that analgesic medications (mu opi-
to take place at arachnoid villi, and the resultant bulk CSF flow was oid, alpha 2 adrenergic, N type calcium channel blockers), aside
thought to influence drug distribution to the spinal cord and brain from the local anesthetics, exert their effects on the target recep-
to a greater extent than by simple molecular diffusion. This bulk tors/channels that are located on the terminals of the primary affer-
flow concept has been shown to be outdated from several perspec- ent and at the level of the first order spinal synapse; and 2) the need
tives, however. for the drug to reach the spinal levels associated with the spinal seg-
Current understandings from imaged-based and computational ments processing the pain information (where the target receptors
fluid dynamics (CFD) show CSF to behave as a poorly mixed volume are located), and the absence of robust infusate redistribution (as
with little net flow, but significant oscillatory flow, originating from discussed in the previous sections). The ability to determine this
CSF pulsations, which are, in turn, influenced by blood pressure, location is, for the most part, difficult, and may be easier to deter-
stroke volume, and intrathoracic pressure variations associated with mine when local anesthetics are administered (174); it is far less clear
respiration. Because of the noncompliant skull, and to a lesser extent when baclofen is infused to treat spasticity or morphine to relieve
spinal canal features, these pulse volumes are transmitted as CSF pain. An important issue is the appreciation that the receptors asso-
pulses, and in the compliant spinal canal lead to oscillatory inflow ciated with the target dermatome are not restricted to the spinal
and outflow at velocities up to 10 mm/sec (167). This pulsatile mix- segment associated with the root dermatome. Afferent input into
ing is orders of magnitude greater that that seen with simple molec- any given segment may send collaterals up to several segments ros-
ular diffusion (164). trally and caudally. It has been argued that the IT drug must accord-
This oscillatory flow interaction with the various IT structures such ingly reach the cells and afferent terminals in these distal
as nerve roots, ligaments, and objects such as catheters induces sec- dermatomes (112,175,176).
ondary flow patterns known as: 1) steady streaming, which may be
more or less than oscillatory flow, but also greater than molecular Consensus Point 23. Limited data exist as to appropriate and
114

diffusion (167), and 2) enhanced diffusion caused by shear forces at best catheter tip placement. The catheter should ideally be centered

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

in the spinal dermatome associated with the pain generator. The Pharmacopeia (USP) chapters Pharmaceutical Compounding—non-
consensus recommendation is that the doctor use clinical judgment sterile and sterile preparations (183). As the FDA does not regulate
based on the clinical setting. these processes, quality assurance is left to the individual pharmacy
or to national compounding associations that offer credentialing.
Pharmacokinetics of IT Analgesic Agents The FDA defines pharmacy compounding as combining, mixing or
Compared to epidural administration, IT administration has long altering of ingredients to create a customized medication for an indi-
been shown to result in higher analgesic efficacy and lower rates of vidual patient in response to a licensed practitioner’s prescription
treatment failures and technical complications (177). The principal (183). If a physician chooses to use a compounding pharmacy, the
advantage of IT therapies involves bypassing the blood-brain barrier. physician should be familiar with quality control procedures of that
This results in higher concentration of administered agents in the pharmacy.
CSF while using lesser amounts of medication. Evidently, greater effi- The USP classifies manipulations of sterile products in aseptic con-
cacy is realized with IT drugs that do not freely cross the blood-brain ditions as low-risk compounding; however, addition of nonsterile
barrier and when the target receptor is predominantly located in the components would constitute high-risk compounding (183). With
CNS in close proximity to the administered IT agent. Medications regard to IT drug delivery (IDD), dilution of commercially available
deposited in the IT space, through catheters placed near the level products such as Infumorph (Baxter Health Care, Deerfield, IL, USA)
processing the patient’s pain, lie close to but not at the target sites. would constitute low-risk compounding, whereas combining an
Except for local anesthetics and baclofen, the receptor sites for IT aseptic product with a powder formulation, such as bupivacaine,
drugs are located in the dorsal horn of the spinal cord, in particular may constitute high-risk compounding. Beyond the quality assur-
in lamina II, also known as substantia gelatinosa (178). In order to ance issues that lie with the individual compounding pharmacy (and
reach their target receptors at neuronal synapses in the superficial are largely out of the control of the prescribing practitioner), there
dorsal horn, intrathecally administered medications must diffuse has been considerable discussion about the role of compounded
across the pia arachnoid and white matter of the spinal cord—a dis- medications for IDD (8,183,184). It should be noted that drug dilu-
tance of up to 1–2 mm from the surface of the cord (179). Diffusion tion is also considered an off-label use of IT medication. This fact,
across the pia is typically considered to be unimpeded given its along with lack of efficacy, has led to the off-label use in the majority
structure of a single layer of cells without intercellular junctions. of IT clinical practice.
The PACC of 2012 commented on the role of compounding, iden-
Work with large molecules has, however, suggested that the diffu-
tifying the risks associated with the practice and outlining basic con-
sion of drugs into the parenchyma may occur though fluid path-
siderations surrounding the practice, such as training of personnel,
ways paralleling the intraparenchymal vasculature (180). The spinal
segregated sterile compounding facilities, air quality of the com-
cord white matter consists of myelinated axons making it hydropho-
pounding area, certification and calibration of equipment, standard-
bic (e.g., dorsal column), whereas the grey matter consists of cell
ized disinfection and quality assurance programs (8). Debate
bodies in the various laminae and is hydrophilic (181). Continuous IT
concerning the practice of using compounded medications contin-
infusion results in stable CSF drug concentrations, which establishes
ues to this day, since essentially all medications could be construed
a gradient driving parenchymal diffusion into the spinal cord. At
as compounded to some degree (185). Around this time, one IDD
equilibrium with slow constant IT infusion, concentrations of small
manufacturer reported that off-label medications or admixtures
molecules in the CSF are thought to be equivalent to those in the
could result in corrosion to the infusion system and device failure
interstitial fluid in the superficial aspect of the dorsal horn (179). (186). This bulletin suggested that preservative-free morphine (maxi-
A number of factors intrinsic to the IT medication play important mum approved concentration 25 mg/mL) was approved for use,
roles in determining drug uptake. Among these, lipid solubility and however, the bulletin also stated that compounded formulations of
molecular weight are the most important physicochemical charac- baclofen and morphine had resulted in motor stall, leading to some
teristics of an intrathecally administered drug. Hydrophilic medica- confusion on the part of practitioners as to what constituted safe
tions administered intrathecally may have a clinical advantage over use of morphine.
hydrophobic or lipophilic IT agents. Compared to lipophilic medica- In 2013, a joint statement by thought leaders from NANS and the
tions, hydrophilic agents have longer half-lives, reflecting the faster American Society of Interventional Pain Physicians presented the
clearance into the vasculature demonstrated by lipophilic agents viewpoint of many physicians experienced in IDD: that medication
(16) and smaller volumes of distribution, resulting in potentially formulations with hydromorphone, fentanyl, and other opioids are
deeper cord penetration and more rostral spread (182). However, more effective than morphine and have fewer side effects (185). In
lipophilic medications have the advantage of limited spread when addition, this statement reconfirms the use of admixtures of bupiva-
precise targeted delivery is desired. caine and clonidine as outlined in the PACC of 2012 (8). The stall
rate for Synchromed pumps (Medtronic Inc., Minneapolis, MN) was
Safety and the Compounding of IT Drugs reported as 2.4% for approved medications (Infumorph, Lioresal,
Medications approved by the FDA have undergone comprehen- and Prialt) at five years and as 4.5% for unapproved medications
sive testing in animal and human subjects to demonstrate safety (185). The PACC suggested that compounded medications were the
and efficacy, while the manufacturing process is continuously evalu- de facto standard of care, and peer-reviewed literature exists to sup-
ated to ensure that high quality standards are met (183). Despite port use of both on- and off-label medications.
these good manufacturing processes and preclinical testing of medi- An infinite number of drug combinations exist and some physi-
cations before FDA approval, there remains considerable need for cians recommend that drug mixtures be utilized only where drug
custom formulations of medications that are not commercially avail- stability information is available. If a study suggests that a high-
able for IT use or commercially unavailable concentrations of medi- concentration drug combination is stable, stability can be assumed
cations. The practice of creating these mixtures of medications, for lower-concentration combinations of the same drugs. For most
known as pharmacy compounding, is not regulated by the FDA but pharmaceuticals, there are established and published standards of
115

rather by state boards of pharmacy, incorporating the United States solubility at room temperature. While high-concentration drug

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

combinations do allow for longer refill interval and delivery of higher revealed an overall success rate of IT opioid therapy of 76.9% of
daily doses, alterations in pH in high drug-concentration solutions patients. In another open-label study, IT morphine was infused in 24
can lead to pump and catheter failure and patient symptoms. For patients with vertebral fractures due to osteoporosis who had not
example, precipitant has reportedly caused catheter obstruction, responded to systemic opioid therapy (198,199). The mean VAS pain
pump corrosion, and failure of IT drug therapy (187,188). score decreased significantly from 8.7 cm before IT therapy to
Given that few clinicians utilize IT medications that are completely 1.9 cm after one year of IT therapy (p < 0.001). Significant improve-
devoid of some pharmacy manipulation (dilution, concentration, ments from baseline to one year were also noted on scores for the
etc.) and that most clinicians utilize compounded off-label medica- Quality of Life Questionnaire of the European Foundation for Osteo-
tions and admixtures, IDD essentially mandates use the of com- porosis subscales for pain, quality of daily life, domestic work, ambu-
pounded medications. Also, since the incident rate of motor stalls lation, and perception of health status (p < 0.001).
for approved and nonapproved medications is similar, it seems pru- In one retrospective study, investigators attempted to determine
dent that diligence in monitoring patients receiving IDD for the pres- characteristics of patients for whom IT morphine therapy is effective
ence of motor stalls or therapy disruption be underscored to detect (200). The study included 131 patients who received IT morphine
and treat these possible outcomes. monotherapy for various pain types (cancer-related, nociceptive, or
neuropathic). A >50% decrease in pain was reported in 73% of all
Review of IT Medications patients. No differences in responder rates were noted when results
To date there has been considerable controversy and little consis- were analyzed by pain type, patient age, or morphine dosage; how-
tency in the trialing of opioids, although trialing with morphine or ever, responder rates were significantly higher in men than in wom-
hydromorphone is common and advised (27,59). The use of pretrial- en (p 5 0.02).
ing systemic opioid dose conversions to derive an appropriate dose Raphael et al. conducted a randomized, double-blind controlled
for IT opioid trialing is not recommended because of pretrial wean- trial of IT morphine efficacy in noncancer pain (201). One group had
ing of systemic opioids and differences in pharmacology between no change in morphine dose while the other had a 20% reduction
systemic and IT opioids. every week for ten weeks. Seven of ten patients, all in the dose-
reduction group, withdrew from the study prematurely. Within-
Morphine group VAS and Oswestry Disability Index differences were statistical-
Mechanism of Action. Morphine is the most widely used IT medi- ly significant between baseline and the last observation for the inter-
cation. It is a mu opioid agonist (189). vention group, with statistically significant greater pain and
worsened disability in the dose-reduction group. These results sug-
Neurotoxicity. Preclinical evaluation in several large animals mod- gest the efficacy of IT morphine for long-term treatment of non-
els showed morphine’s propensity to initiate space occupying cancer pain.
masses or IT granulomas (190–193). These masses, constituted of
fibroblasts, maturing collagen and interspersed with inflammatory Hydromorphone
cells, arise from the dura/arachnoid, with the mass size largest proxi- Mechanism of Action. Hydromorphone is a mu opioid agonist
mal to the catheter tip (188,194). This profile has been observed (202).
with several opiates, including hydromorphone and methadone
(131). The association between IT opioid therapy and granulomas is Neurotoxicity. Preclinical studies of IT infusion of hydromorphone
further discussed in a PACC 2012 publication (60). in large animal models showed space occupying granulomas at
higher concentrations (131,203).
Clinical Studies. Clinical data on IT morphine continue to support
its use as a first-line therapy. From 1983 to 2000, there were many Clinical Studies. The literature review revealed no new studies
studies showing efficacy of the long-term infusion of morphine and investigating the efficacy of IT hydromorphone in the treatment of
morphine/adjuvant admixtures, as reported by PACC 2012 (8). chronic pain. Two case reports described granuloma development
Recent results from several long-term studies support the efficacy of in patients treated with IT hydromorphone. The first described a
IT morphine in treating patients with chronic pain, including pain patient who developed a granuloma on IT morphine; nine months
from cancer and noncancer diseases. In a retrospective study, medi- after removal of the first granuloma, she developed another granu-
cal records from 57 patients with chronic malignant pain on long- loma after one month of IT hydromorphone therapy (204). The sec-
term IT opioid therapy (morphine, hydromorphone, or sufentanil) ond report described a 52-year-old man with a history of chronic
were reviewed (195). VAS scores for pain significantly decreased lumbar spine pain who developed a granuloma while receiving
from baseline to time of first refill (p  0.001); VAS scores then high-concentration IT hydromorphone (85 mg/mL) at a dose of
remained stable and significantly lower than baseline scores 19.8 mg/d (205).
(p  0.001) through year 3. Oral opioid use decreased significantly in Mallinkrodt Phamraceuticals (St. Louis, MO, USA) is enrolling
the first year of IT therapy (p  0.001) and increased slightly but patients in a study to develop a branded and formally manufac-
insignificantly between years 1 and 3. tured, FDA-approved hydromorphone product. The first trial is a
In a prospective, open-label study of IT morphine infusion (Prom- controlled, two-arm, parallel-group, randomized withdrawal study.
etraVR Infusion Pump, Flowonix Inc., Mt. Olive, NJ, USA), 110 patients Subjects in this trial will already have implanted IT pumps and will
with chronic pain were treated and followed up for approximately be transitioned to IT hydromorphone. They will then be titrated to a
one year (196). Pain relief was noted within one month and was sus- level where oral opiate medications are eliminated up to a dose of
tained during the following six months; trends indicated consistent 5 mg of IT hydromorphone per day. Subjects who attain stabilization
pain relief through 12 months. In an open-label study, 13 patients and meet criteria for randomization will be assigned to either remain
with intractable pain from chronic pancreatitis who had undergone at their current dose of hydromorphone or be titrated off therapy in
a successful trial of IT opioids received IT opioid infusions for a mean a blinded fashion. The primary efficacy end point of this study is the
116

duration of 29 months (197). The limited intention-to-treat analysis proportion of subjects who are treatment failures during the

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

double-blind randomized withdrawal period. The second trial is a Neurotoxicity. IT ketamine infusion (10 mg/mL delivered at 2.4 mL/
Phase 3, open-label, single-arm safety study where subjects can d) in chronically catheterized dogs resulted in mild to severe spinal
either directly enter the study from the previously described trial or pathology ranging from local demyelination to necrotizing lesions
be transitioned directly to IT hydromorphone. The two formulations of spinal parenchyma near the catheter tip (10). Similar pathology
of IT hydromorphone being investigated are a 2 mg per cc and a was observed in neonatal rats (213). This effect was shared by other
10 mg per cc concentration, respectively. These trials are actively N-methyl-D-aspartate (NMDA) antagonists, including MK801, mem-
recruiting subjects and results are expected in 2017. antine, amitriptyline, and S-methadone. Notably, these studies
Peripheral edema associated with IT hydromorphone infusion was unfortunately did not establish a no effect level nor correlate the
reported in one patient. This 61-year-old woman with chronic pain lower doses with an antihyperpathic action.
developed progressive lower extremity edema, which was compli-
cated by severe cellulitis, while on IT morphine (206). Her edema Clinical Studies. A randomized, double-blind study compared the
lessened when she was switched to IT hydromorphone but recurred use of epidural ketamine plus bupivacaine vs. epidural bupivacaine
with severe cellulitis two months later. Her IT regimen was changed plus saline in 53 patients undergoing lower limb amputation (214).
to clonidine (33 mcg/d) and baclofen (67 mcg/d); edema resolved In both treatment groups, persistent phantom and stump pain were
and did not recur. less than that seen in comparable studies and did not differ signifi-
cantly between groups. In the ketamine/bupivacaine group, signifi-
Fentanyl cant decreases from preoperative anxiety and depression levels
Mechanism of Action. Fentanyl is a lipophilic mu opioid agonist were noted and persisted through the one-year follow-up point.
(132,207). Additionally, a case report described a 49-year-old woman with
severe cancer-related upper back and abdominal pain (215). Her
Neurotoxicity. Preclinical studies of IT infusion of fentanyl or alfen- numeric rating scale (NRS) pain score was 6, despite 96 days of IT
tanil in large animal models showed no space occupying granulo- therapy with a combination of morphine and bupivacaine. IT keta-
mas at the highest concentrations examined (132,133). mine was added to her regimen and her NRS score decreased to 3.
There were no signs of motor paralysis, psychomimetic alteration,
Clinical Studies. The literature review revealed no new studies neurological dysfunction, or infection in this terminally ill patient.
investigating the efficacy of IT fentanyl in the treatment of chronic This contrasts with subpial vacuolar myelopathy, which was found
pain. One case report described a 34-year-old woman receiving IT postmortem in a cancer patient treated with 5 mg/day IT ketamine
combination therapy (fentanyl, bupivacaine, and clonidine) for for three weeks (216).
chronic pain who was suspected of having an epidural hematoma
because of inadequate pain control despite increasing doses and an
Methadone
unsuccessful epidural steroid injection (208). On operation, a
Mechanism of Action. Methadone is a racemic compound in which
catheter-tip mass was noted within the epidural space, with the
the d-isomer has NMDA receptor antagonist activity and the l-
catheter tip in its center. Misplacement of the catheter (epidurally
isomer is a mu opioid agonist (189).
instead of intrathecally) at the time of original insertion complicated
diagnosis. Histopathological analysis revealed a proteinaceous mass,
Neurotoxicity. Notably, there is concern about the safety of IT
which the authors determined was an inflammatory mass, not a
methadone, since all compounds with NMDA activity have serious
granuloma, and was likely a result of drug precipitation.
neurotoxic effects (212). Continuous infusion of the isomers in a dog
model revealed spinal toxicity and granulomas with either isomer
Sufentanil
(131).
Mechanism of Action. Sufentanil is a potent mu opioid agonist
(209).
Clinical Studies. The efficacy of epidural methadone was investi-
gated in a study of 32 patients with cancer-related pain that was
Neurotoxicity. No canine continuous infusion trials with sufentanil
refractory to epidural morphine (217). Patients received one of the
have been reported. Repeated bolus delivery in the canine mode
following treatments: 2.5, 5, or 7.5 mg epidural methadone diluted
showed no histopathologic changes (132).
in 60 mg lidocaine or 7.5 mg epidural methadone diluted in 60 mg
lidocaine plus 10 mg dexamethasone. Epidural methadone provided
Clinical Studies. The literature review revealed no new studies
dose-dependent analgesic effects, and these effects were further
investigating the efficacy of IT sufentanil in the treatment of chronic
improved with the addition of dexamethasone. A prospective study
pain. One case report described an 86-year-old woman with FBSS
of IT methadone was performed in 24 patients (218). Thirteen
who had received multiple IT therapies over the course of two years
patients experienced improvement of their pain control with metha-
(210). Six weeks after beginning IT sufentanil therapy (12–17.2 mcg/
done, nine continued to receive this agent for six months with good
d), she had lower extremity weakness, sensory changes, and intracta-
pain relief.
ble lumbar pain, and a CT-myelogram demonstrated the presence
of a granuloma. Sufentanil was removed from the pump and
replaced with normal saline. Her symptoms resolved within approxi- Ziconotide
mately 48 hours, and the patient was receiving oral methadone ther- Mechanism of Action. IT ziconotide is first-line therapy for both
apy for pain at the time of hospital discharge. neuropathic and nociceptive pain, and is FDA approved. Its mecha-
nism of spinal action is to block presynaptic N-type calcium channels
Ketamine in the dorsal horn of the spinal cord (219,220). This targeting is dis-
Mechanism of Action. Ketamine is a noncompetitive antagonist tinctly different from mu agonism and allows ziconotide to be help-
117

that blocks the glutamate NMDA ionophore (211). ful in the opioid-tolerant patient (32).

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

Table 24. Recommendations Regarding Intrathecal Clonidine Treatment by the PACC Using USPSTF Criteria.

Statements Evidence levels Recommendation strength Consensus strength

Intrathecal clonidine in CRPS patients decreases pain scores over time as I A Strong
well as allodynia, hyperalgesia, and mean arterial blood pressure.
Clonidine increases analgesia duration and decreases morphine use in the II-2 B Strong
acute postoperative setting.
Clonidine may precipitate hypotension in patients with baseline II-3 B Strong
hypertension.
Ziconotide concentration decreases over time when mixed with clonidine. II-3 B Strong

Neurotoxicity. Ziconotide (Prialt) has undergone extensive preclini- Clinical Studies. Though not FDA-approved for continuous IT use,
cal safety evaluation in multiple species (221) without spinal toxicity bupivacaine is the most common local anesthetic used in spinal
in the concentrations employed. anesthesia and is used off-label in IT therapy. Compared to the epi-
dural route, IT drug delivery results in higher patient satisfaction,
Clinical Studies. Many methods have been employed to trial zico- fewer catheter complications, better pain relief and sleep (174).
notide, from continuous infusion to single-shot bolus. There is no Combinations of bupivacaine and opioids have shown synergistic
evidence to suggest one trialing strategy is better than another, efficacy in acute postoperative and labor pain studies (110,236–239).
appreciating that on-label FDA trialing is performed by a microam- In chronic pain settings with continuous IT drug delivery, howev-
bulatory delivery device, or via the Medtronic Synchromed II device. er, the effect is less clear. A retrospective study of 109 patients, who
In patients with neuropathic pain who are not responsive to pre- were managed with a solution containing a mixture of IT bupiva-
trialing systemic opioid therapy, a trial of ziconotide should be con- caine and opioids after an initial period of IT opioid-only treatment,
sidered. Trialing with ziconotide can be challenging because the found improved pain control and decreased oral opioid consump-
drug’s narrow therapeutic window and the side effect profile is tion with combination therapy compared to opioids alone (134).
more closely related to the rate of dosage increase. Thus, trialing The average daily bupivacaine dose in that study was 10 mg. A small
with an externalized catheter may be impractical and hazardous double-blind randomized prospective study suggested that addition
because of the slow titration required with ziconotide (e.g., dosage of bupivacaine (up to 8 mg/day) to IT pumps infusing morphine or
increases of 0.5–1.0 mcg every several days). Although trialing with hydromorphone did not result in improved pain control in patients
bolus dosing can be useful to identify some appropriate candidates, with low back pain, mostly in the setting of postlaminectomy syn-
side effects associated with bolus dosing may eliminate many drome (172). However, a large study in noncancer pain patients
patients who might otherwise have benefited from IT ziconotide revealed blunting of opioid dose escalation in IT-therapy patients
therapy. Thus, the use of alternative trialing methods in order to receiving bupivacaine in addition to IT opioids. The average bupiva-
avoid a trial failure because of intolerable side effects would be caine daily dose in those patients was 9.8 mg at one year postim-
advantageous in this regard. Although meclizine treatment is some- plant (99). Nevertheless, the difference in pain scores between the
times used before IT ziconotide trials in clinical practice, there is group receiving IT opioids and the group receiving IT opioids plus
insufficient evidence to support this approach. Proper hydration via bupivacaine was not statistically significant. A similar effect of blunt-
ing IT morphine dose escalation was noted in a small cancer pain
intravenous (IV) infusion before trialing may limit the side effect of
study (240).
hypotension.
The high lipid solubility of bupivacaine limits its spread intrathe-
cally and highlights the need to place the catheter in the posterior
Bupivacaine IT space at the site of processing-pain pathology (20). No formal
Mechanism of Action. Bupivacaine is an amide local anesthetic studies have been performed to assess starting and maximal doses
with high lipid solubility that is often used off-label in IT therapy of bupivacaine in IT therapy. In addition, there are no prospective
(99,134,222,223). Several mechanistic properties give it utility with studies of chronic IT bupivacaine administration as a sole agent. The
spinal delivery: 1) differential efficacy at low concentrations that most recent PACC guidelines have suggested a maximal concentra-
alters sensory processing while sparing motor function (224–227); 2) tion of 30 mg/mL, a starting dose of 1–4 mg/day and a maximal dai-
absence of tachyphylaxis in patients with neuropathic or somatic ly dose of 10 mg (8). However, this maximal dose is similar to the
pain (174,228–230); and 3) potent synergy with other IT analgesic average dose noted to be effective in previous studies (99,134).
targets in animal models (110,231) and in humans (226,227,229,230). Additional bupivacaine is sometimes self-administered in boluses by
patients through a personal therapy manager (PTM) device (241).
Neurotoxicity. In early rat studies with continuous bupivacaine Serious cardiotoxic side effects can occur when significant amounts
infusion, modest increases in neuronal vacuolation, was observed at of bupivacaine reach the bloodstream. This should not be of con-
concentrations of 0.5% (232). Bolus delivery in dogs of bupivacaine cern with IT bupivacaine infusion (224). Clearly, the limiting factor in
(0.75%) resulted in minor leptomeningeal cellular infiltration (233). IT bupivacaine infusions would be sensorimotor loss. Nevertheless, IT
infusion of 2.5–3.8 mL/d of 0.25% bupivacaine for 3–11 weeks bupivacaine doses as high 5 mg/hour (or 120 mg/day) and bolus
resulted in mild leptomeningeal cellular infiltration in two of eight doses as high as 7.0 mg in the high cervical IT space have been
animals (233). In combination with morphine in human cancer reported with no apparent untoward manifestations (242). Catheter
patients no significant histopathology was noted on autopsy tip location, CSF dynamics, and patient mobility likely play important
(234,235). IT infusion of bupivacaine has a long track record of safety roles in sensorimotor loss in response to bupivacaine. The most
118

and efficacy alone or in combination with morphine. recent version of PACC guidelines suggested that, in neuropathic

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Table 25. Recommendations Regarding Intrathecal Baclofen Treatment by the PACC Using USPSTF Criteria.

Statement Evidence level Recommendation grade Consensus strength

Baclofen should be considered an intrathecal medication for use to treat spasticity. II-2 A Strong
Baclofen can be used as an adjuvant to treat pain. II-3 B Moderate
Care regarding mitigating withdrawal from baclofen is suggested. II-2 A Strong
Ancillary resources regarding physical therapy to aid in titration and assessment when III C Moderate
employing baclofen is recommended.
Using bolus or flex dosing strategies to improve spasticity demonstrates promise. II-3 B Moderate

pain, bupivacaine in combination with morphine is considered first antihyperalgesic effects at the spinal level (251,252). Concurrent
line but second line in combination with morphine, fentanyl or with these effects, baclofen at the GABA-A receptor can have promi-
hydromorphone in nociceptive pain (8). It should be noted that nent effects on motor tone via direct hyperpolarization of the motor
there is no basis for such an assertion. Recent data suggest efficacy horn cells.
of IT bupivacaine in combination with hydromorphone as first-line
therapy (135). Average daily dose of bupivacaine at 24-month post- Neurotoxicity. IT baclofen infusion (at rates of up to 2 mg/mL/d)
implant was 12.1 6 0.9 mg including on average 3.7 6 0.6 PTM for 28 days in chronically catheterized dogs has been shown to
boluses of 0.78 6 0.05 mg bupivacaine each. result in no behavioral or spinal histological evidence of neurotoxici-
ty (253). Additionally, preclinical evaluation suggested that IT baclo-
Clonidine fen at doses up to 2 mg/mL/d were not associated with granulomas
Mechanism of Action. Clonidine is an alpha2 adrenergic agonist
in dogs (254). The development of granulomas in humans is rare
(243). Clonidine may exert antiallodynic effects by inhibiting the acti-
with IT baclofen therapy (255). Granuloma formation was reported
vation of glial cells and by activation of nuclear factor jB and p38
in two patients receiving IT baclofen monotherapy (256). However,
(MAP kinase), thus inhibiting the production of proinflammatory
these reports, plus another, were later re-evaluated, and other scien-
cytokines (244). Increasing evidence suggests that activated spinal
tifically plausible explanations (e.g., baclofen precipitation) were
cord glial cells contribute to enhanced pain states through the
posited for MRI findings in these patients who were originally
release of proinflammatory cytokines, such as tumor necrosis factor-
reported to have IT baclofen-induced granulomas (257). The associa-
a (TNF a), IL-1, and IL-6 (245,246).
tion between IT baclofen therapy and granulomas is further dis-
cussed in the brief report titled, “Polyanalgesic Consensus
Neurotoxicity. Results of studies in large animals treated with epi-
Conference 2 2012: Consensus on the Diagnosis, Detection, and
dural clonidine for 28 days (concentrations up to 2 mg/mL and
Treatment of Catheter-Tip Inflammatory Masses (Granulomas)” (60).
doses up to 7.7 mg/d) revealed no notable histopathologic findings
(245). Additionally, IT infusion of clonidine (2 mg/mL at 2.4 mL/d)
Clinical Studies
monotherapy for 28 days was not associated with direct evidence of
Neuropathic Pain. Recent reports of the use of IT baclofen for the
spinal histopathology (191). In the same study, in dogs that received
treatment of patients with neuropathic pain include two studies and
admixtures of clonidine and morphine, the severity of spinal histopa-
two case reports. In a double-blind study, the effect of different IT
thology decreased in a clonidine dose-dependent manner.
baclofen infusion rates (i.e., 0.75 or 3 mg/mL baclofen solution
infused at a consistent rate) on pain and dystonia was investigated
Clinical Studies. Combination therapy including IT clonidine was
in 14 patients with complex regional pain syndrome (CRPS) who
described in a case report of a 79-year-old man with chronic lower
had not responded adequately to previous IT baclofen therapy
extremity pain (246). Approximately one year after beginning IT
(258). Overall, the faster baclofen infusion rate was not associated
therapy with fentanyl, bupivacaine, and clonidine, the patient
with improvements in dystonia or pain but was associated with
reported night terrors, insomnia, severe dry mouth, and increased
increased frequency of adverse events (AEs). However, in a subset of
depression. Three days after discontinuation of clonidine therapy,
six patients for whom AEs had previously prohibited dose escalation
his depression improved and the other symptoms resolved; the
of IT baclofen, all but one preferred the faster infusion rate, reporting
symptoms have not recurred after >2 years of clonidine-free IT ther-
that the effects of the faster-infusion IT baclofen on pain and dysto-
apy. Clonidine has been evaluated in many studies, with improve-
nia outweighed the severity of AEs. One report described two cases
ment in analgesia and opioid-mitigating effects (247–250).
The PACC recommendations for IT clonidine appear in Table 24. of baclofen and ziconotide combination therapy (259). The first
patient was a 48-year-old man with neuropathic pain who had
Baclofen received ziconotide (2.4 mcg/d) for approximately three months
Baclofen is commonly used for intractable spasticity and is FDA before baclofen (110–115 mcg/d) was added to his IT regimen for
approved for use in IT pumps. Baclofen has limited use as a mono- spasticity control. His ziconotide dosage was then reduced to 1.7
therapy option for the primary treatment of chronic pain. It is used mcg/d over the course of one month. After eight months of zicono-
most commonly in combination therapy to treat pain with spasticity tide/baclofen therapy, his VASPI score had decreased by 75%. The
(Table 25). second patient was a 73-year-old man with neuropathic pain who
had received ziconotide monotherapy (dosage at onset of pain
Mechanism of Action. Baclofen is an agonist of the gamma- relief, 14.4 mcg/d) for six months when baclofen (62 mcg/d) was
aminobutyric acid (GABA)-A receptor. In preclinical studies, the added for control of spasticity. After two years on ziconotide/baclo-
119

GABA-A receptor, a chloride ionophore, has been shown to exert fen therapy, his VASPI score had improved from baseline by 30%. He

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

Table 26. Intrathecal Drug Delivery Systems.

Codman 3000 Medtronic Isomed Flowonix Prometra II Medtronic SynchroMed II

Catheter material Polyurethane with titanium Radiopaque silicone rubber Radiopaque silicone rubber Radiopaque silicone rubber
reinforced coil with titanium tip with tungsten tip with titanium tip
Pump material Titanium/silicone rubber Titanium/silicone rubber Titanium/silicone rubber Titanium/silicone rubber
Pump mechanics Continuous flow propellant Continuous flow propellant Valve gated programmable Peristaltic titanium/
plastic programmable
MRI compatibility No effects 3T No effects 1.5T MRI conditional 1.5T MRI conditional 3T
with valve shut-off
Patient-controlled intrathecal None None Patient therapy Personal therapy
analgesia (PCITA) controller (PTC) manager (PTM)

also experienced improvements in mood and ability to perform hypotonia, seizures, autonomic instability, bradycardia, and respira-
activities of daily living during this time. tory depression (269). One case report described baclofen overdose
associated with a change in IT baclofen concentration combined
Trialing. In a study that included 48 patients with neuropathic pain with the performance of a catheter dye study (269).
who had inadequate response to SCS, participants were given IT bac-
lofen boluses (25–100 mcg) (260). Among these patients, 14 were Combinations of IT Drugs
classified as responders (>50% improvement from baseline in pain A number of studies have been conducted to evaluate the use of
level), and 11 had pumps implanted for continuous IT baclofen infu- IT morphine in combination with other IT agents, such as bupiva-
sion (four with pumps alone, seven with SCS plus pumps). Follow-up caine, ziconotide, and baclofen. One such open-label study included
after an average of 32 and 67 months of SCS plus baclofen therapy 55 patients with advanced cancer-related pain who had been unre-
revealed that >50% of patients maintained good treatment effects; sponsive to previous trials of systemic opioids alone and were
baclofen doses approximately doubled during this time. treated with a combination of IT morphine and IT bupivacaine and
followed for up to six months (270). The initial IT morphine dosage
Tolerance. Tolerance/tachyphylaxis is an important consideration was calculated from the patients’ previous systemic opioid dosage
when using IT baclofen, as it may occur in approximately 22% of by using an oral:IT ratio of 100:1 (which is notably different from the
patients treated with long-term IT baclofen (261). Tolerance may 300:1 ratio that is typically used for equianalgesic calculations) (271).
develop even after very long-term treatment, as was described in a The initial bupivacaine dosage of 12.5 mg/d was increased to
case report of a patient who developed tolerance 16 years after initi- 25 mg/d before the IT morphine dosage was increased and modi-
ation of IT baclofen therapy (262). A drug holiday of 24 hours fied as needed. Significant reductions in pain intensity, along with
(with careful monitoring for withdrawal symptoms) may be helpful. significant decreases in the mean systemic opioid dose, were noted
Additionally, limited data in four patients suggest switching to a pul- at one and three months after initiation of IT therapy and up to the
satile bolus infusion may help address tolerance (261). time of death (p  0.029). In another open-label study, which includ-
ed 32 patients with chronic noncancer pain who had >70% pain
Withdrawal. Abrupt cessation of IT baclofen therapy could result in relief after a trial of low-dose IT morphine and bupivacaine, continu-
baclofen withdrawal, a serious, life-threatening situation that can be ous IT therapy (0.1 mg/d morphine, 0.5 mg/d bupivacaine) was initi-
severe and prolonged (263). Baclofen withdrawal may mimic seroto- ated, and dosages were titrated to a mean of 1.03 mg/d morphine
nin syndrome (264) and has rarely been associated with hallucina- and 1.15 mg/d bupivacaine (272). Mean VAS pain scores decreased
tions (265). One case report described baclofen withdrawal after significantly from baseline to month 3 (p < 0.01) and remained con-
removal of an IT baclofen pump in a 45-year-old woman with para- sistently reduced through the 48-month follow-up.
plegia and severe lower extremity spasticity (266). She was treated The addition of IT morphine in 25 patients with suboptimal pain
with oral baclofen, lorazepam, phenytoin, and tizanidine and gradual- relief on stable dosages of IT ziconotide was investigated in an
ly improved over the course of seven days. She was discharged on open-label study (273). VASPI scores for these patients improved by
phenytoin, linezolid, and metoprolol, with no need for oral spasticity a mean of 26.3% by week 4 of combination therapy, and mean sys-
therapy. It is also important to note that IT baclofen withdrawal may temic opioid consumption decreased by 49.1%. Notably, stability
result from catheter leakage (267). Clinicians should be aware of the data regarding ziconotide and opioid admixtures may provide guid-
signs and symptoms of baclofen withdrawal and be watchful for ance for frequency of pump refills (115,274).
them in any patient who receives IT baclofen. One report described
the successful weaning of a patient from high-dose IT baclofen thera- INTRATHECAL DRUG DELIVERY SYSTEM
py through use of a lumbar drain and standard PCA pump delivering
continuous infusion of IT baclofen as a means of avoiding withdrawal CHARACTERISTICS AFFECTING IT THERAPY
(268). It should also be noted that symptoms of baclofen withdrawal Pump and Catheter Materials and Mechanics
might be the first indication of IT catheter migration. Since baclofen Intrathecal pumps can be mainly differentiated into systems that
is a water-soluble agent, migration of the catheter into the epidural are continuous flow or variable flow. The driving mechanisms may
space will result in symptoms of baclofen withdrawal. include peristalsis, fluorocarbon propellant, osmotic pressure, piezo-
electric disk benders, or the combination of osmotic pressure with
Overdose. Baclofen overdose is a potentially life-threatening condi- an oscillating piston (Table 26). Pump materials are similar with the
120

tion, the signs and symptoms of which may include somnolence, pump shell being titanium and filling ports containing silicone

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

rubber. Physical orientation of the filling and side ports are largely biophysiologic properties that determine drug distribution in the
consistent, with differences in negative pressure or positive pressure spinal canal are incompletely understood. Many factors have been
confirmation strategies (275). proposed and evaluated as contributing to differential drug distribu-
Pump delivery mechanics include continuous flow propellant or tion in the CSF. For instance, anatomic variation, postural changes,
programmable features. Propellant pumps (Codman 3000 and Med- drug solution density, binding characteristics of drugs at the dorsal
tronic Isomed) do not require batteries and deliver a continuous horn, CSF volume and variations in CSF pulsatile flow with heart
flow for the life of the pump. The programmable pumps require bat- rate, stroke volume, and respiratory cycle have been examined
tery replacement, based on labeling, at a maximum of five to seven (20,156,170,281,283). Additionally, it has been suggested that the
years for the Medtronic Synchromed II and maximum of 10 years for rate of dispersion in the CSF cannot be explained by diffusion alone
the Flowonix Prometra II. (156,283).
The programmable pump systems feature differences that deserve CSF convective transport within the spinal canal has been studied
mention. The Medtronic Synchromed II (Minneapolis, MN, USA) sys- exclusively, and it is now known that CSF flow is pulsatile with oscil-
tem uses a peristaltic rotor system of internal tubing to deliver medi- latory displacements creating microenvironments with eddy cur-
cation from the reservoir to the external catheter system. The rents resulting in complex micromixing of infused drugs with no net
Prometra II Flowonix Pump (Mount Olive, NJ, USA) employs a valve- bulk flow (20,156,281,283). A recent study based on computer
gated bellow delivery mechanism. Each pump has the ability for modeling of microanatomic structures in the spinal canal suggested
patients to deliver patient-controlled dosing by using a patient-held that the spinal nerve roots themselves serve as a significant barrier
programmer (Patient Therapy Manager or PTM for Medtronic and to laminar flow and may create much of the geometric-induced
the Patient Therapy Controller or PTC by Flowonix). Both pumps sup- flow patterns observed and postulated through various experimen-
port MRI conditional labeling, with the Medtronic pump up to 3 Tes- tal designs (281). With regard to rostral-caudal spread of hydrophilic
la and the Prometra pump at 1.5 Tesla. Of note, both pumps require medications such as morphine, it has been demonstrated that a
interrogation following a scan. For the Medtronic system, exposure rather steep concentration gradient exists as samples are taken at
to a magnetic field will create a motor stall, which typically resolves points further removed from the catheter tip. The authors of this
following removal of the magnet and can occur within 20 min to 2 finding suggested that drug dilution over distance and drug concen-
hours, with a failure of motor stall recovery on rare occasions. For tration at the site of action may be important in providing analgesic
this reason, it is suggested to interrogate after the scan (276). efficacy (170). Given these data, simplistic views of CSF dynamics do
The Flowonix Prometra I system requires removal of all medica- not provide insight into the possible clinical implications of decisions
tion within the reservoir prior to MRI exposure, as failure can result made surrounding drug-infusion rate and drug dispersal within the
in emptying of the reservoir contents into the patient. The Prometra CSF. Much of what is known clinically is derived from observations
II system remedied this concern with the flow activated valve (FAV) utilizing spinal anesthesia for surgical intervention (282). Despite this
that is triggered when exposed to a magnetic field, blocking drug complexity there are some basic observations that have been
delivery from the reservoir to the patient after delivery of less than reported in the literature that provide insight into the clinical utility
or equal to 10 lL (275). If the contents of the reservoir are expected of IT drug delivery flow-rate manipulations on treatment outcomes.
to be less than 1 mL, they should be removed prior to the MRI Basic Science
because the FAV may not activate. After the MRI, the contents of the Detailed examinations of the effects of the flow rate in IT drug
reservoir have to be removed entirety to manually reset the FAV, delivery have been conducted by the late C. Bernards in a porcine
the pump interrogated and the contents replaced in sterile fashion, model (20). This model has a number of advantages over previous
with elapsed time of 3 min (275). models in that it mimics IT therapy, namely a closed model with no
The Medtronic Synchromed II System has a minimal flow rate of CSF loss or disturbance due to sampling, preservation of the effect
0.048 mL/day to allow for programming, while the Prometra II sys- of cardiac and respiratory cycles, an upright position and the ability
tem can be at zero flow. Accuracy of the Prometra system is greater to study drug concentrations in both spinal fluid and spinal cord.
(97.8%) compared to the Medtronic Synchromed II system (2 vs. Bernards compared flow rates of 20–1000 mcL/hour and a bolus
14.5%) (276–279). The Medtronic system has two reservoir sizes, 20 group receiving a bolus of 1000 mcL/hour administered more than
and 40 mL, while the Prometra II system has a 20 mL reservoir only. 5 min every hour (20). These rates were chosen to be representative
Although it is beyond the scope of the PACC, the consensus of regular and maximum clinical pump flow rate as well maximum
group felt it necessary to comment briefly on the warning letters speeds achieved by bolus administration. The most prominent find-
surrounding the Medtronic Synchromed II system, and the recent ing was the limited distribution of bupivacaine and baclofen from
consent decree agreement between the FDA and Medtronic in April the site of administration, especially in the 20 mcL/hour group. For
2015. Prior to this, warning letters were released regarding overinfu- both bupivacaine and baclofen, most of the drug recovered in the
sion, corrosion of the internal tubing with the use of off-label medi- CSF and spinal cord in this group was found within 1 cm of the site
cations or combinations or medications, and priming bolus errors of administration. Diffusion of both bupivacaine and baclofen in CSF
(280). The complexity this introduces for use of the Medtronic Syn- or spinal cord parenchyma was increased in the 1000 mcL/hour and
chromed II system is unknown, with a recent editorial offering a bolus groups compared with the 20 mcL/hour group. Evidence of
foundation for discussion (5). greater distribution comes from the dose-normalized CSF area under
the curve and spinal cord concentration data. Evidence that the
Intrathecal Infusion Rate bolus group achieved better drug distribution than did the 1000-
Intrathecal therapy offers advantages over systemic therapy in mcL/hour group was more subtle but still present. Bernards con-
that IT delivery bypasses the blood-brain barrier with direct access cluded that CSF is a poorly mixed medium, that CSF motion is
of the drug delivered to receptor sites in the dorsal horn of the spi- limited and that the spread of drug molecules in CSF is largely
nal cord (283). The efficacy of this therapy is dependent to some dependent on the kinetic energy imparted to the drug molecules by
121

degree on drug distribution within the spinal canal; however, the the infusing mechanism. The clinical implications of Bernards’ work

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

Table 27. Recommendations Regarding Infusion Rate by the PACC Using USPSTF Criteria.

Statements Evidence levels Recommendation strength Consensus strength

Rate of dispersion in cerebrospinal fluid cannot be attributed to diffusion alone. II-2 B Strong
Flow rate may not impact analgesia. II-2 C Weak
Bolus dosing may improve analgesia. II-2, II-3 B Strong

are that the location of the tip of the catheter relative to the tar- satisfaction and reduced need for oral medication supplementation
geted spinal cord segment may be critical given the limited capacity (241). Recently, it has been reported that patient-controlled IT anal-
for CSF to distribute drugs away from the catheter tip. Limited distri- gesia with bolus dosing results in better patient satisfaction in
bution of morphine away from the catheter tip may predispose to cancer-related pain (282). No further work on bolus dosing with IDD
formation of IT granulomas, and development of methods to has been done to our knowledge (283–285).
improve drug distribution may decrease the risk of granuloma All told, these data suggest a lack of benefit from increasing infu-
formation. sion rate and, in fact, there may be a clinically significant deleterious
Flack et al. went on to confirm these findings in a chronic ambula- effect, as decreased quality of life has been reported with increasing
tory pig model receiving IT morphine over a period of 14 days (90). flow rate (283). The one preliminary report using intermittent bolus
In this experiment, four pigs were implanted with IDD systems and dosing in addition to constant infusion suggests a positive impact
an infusion of morphine delivered at 20 mcL/hour. The authors con- on patient outcomes but has not been replicated. Reck et al. dem-
cluded that the chronic data confirmation of limited CSF distribution onstrated, in a blinded crossover study of ten patients comparing
in the ambulatory animal may help to explain clinical situations in bolus to continuous infusion, a statistically significant reduction of
which a drug delivered as an IT bolus at trial stage is effective at numerical rating scores with intermittent, programmed, bolus deliv-
relieving patient symptoms but, when delivered at the very slow ery, compared to continuous infusion (286). No conclusions regard-
infusion rates used for chronic infusion, was not effective (i.e., phar- ing safety and efficacy can be drawn from the limited data currently
macokinetic failures of chronic IT drug delivery). Specifically, published. However, with the technology now available making it
although bolus IT injection produces relatively widespread drug dis- possible for bolus therapy in multiple applications to be provided to
tribution, that was not the case with chronic infusions. patients, this therapy is being utilized by increasing numbers of pro-
viders and data will be forthcoming. Caution and a conservative
Clinical Studies approach is advised when choosing to utilize intrathecal bolus ther-
Two clinical studies have examined the effect of varying the infu- apy as our understanding of flow dynamics, oscillatory mechanisms
sion rate for baclofen and varied mixtures of analgesics, respectively. and drug bioavailability are still evolving.
Both studies utilized a similar double-blind crossover design over The PACC recommendations for infusion rate appear in Table 27.
two-week periods with a constant daily drug dosage throughout
but an infusion rate varying at random from the patient’s baseline Baseline Dose of Opioids: High vs. Low or None
rate to twice and four times this value. In the baclofen study van der The impact of oral opioid therapy on subjects trialed and
Plas et al. (258) randomized patients who experienced no beneficial implanted for IT therapy has been examined in several recent stud-
response or excessive side effects to intrathecal baclofen (ITB) infu- ies (14,15,100,198,284,285). The techniques surrounding manage-
sion for dystonia to either slower infusion rate delivery or four-times ment of oral opioid therapy in those considering IDD range from
faster infusion rate delivery (FIRD) for two weeks. Patients crossed leaving the patient on oral opioids and adding IDD to pretrial/
over after a one-week washout period. The authors observed no sig- implant taper of opioid medications. Anderson et.al. in 2003
nificant differences between the two groups for the median change reported outcomes after taking subjects off opioids 12 hours before
of numeric rating scale dystonia (20.3 [interquartile range {IQR} the trialing period (284). This was followed by several case studies
21.1 to 0.5]), pain (0.1 [IQR 20.8 to 1.3]). However, they found that that described various methods of tapering mediations during the
the frequency of AEs was significantly higher during FIRD (12 vs. 2). trialing period (101,195). Shaladi et al. (198) lowered oral opioid
Only patients who were included because side effects to ITB pre- doses as IT doses increased during the trial, while Kim et al. (100) dis-
vented dose escalation preferred FIRD. Investigators concluded that continued opioids 4–12 hours prior to trialing.
given a fixed daily dose, a four-times higher infusion rate enhances Subsequent larger studies examined the role of eliminating oral
the IT distribution of baclofen as evidenced by the significantly opioids and the effects on the efficacy of IDD (14,15). Grider et al. in
higher number of AEs. However, in CRPS a fourfold higher infusion a small case study and later in a larger retrospective study reported
rate was not associated with clinically overt improvement of dysto- discontinuing oral opioids for six weeks prior to trial/implant. In that
nia or pain. Patients in whom side effects restricted further dose study the pretrial VAS score on oral opioids was compared to the
escalations of ITB favored the faster rate because of subjective VAS following opioid taper and six weeks in an opioid-free state,
improvement of dystonia and pain. Therefore, the utility of a faster demonstrating that patient-recorded VAS scores were virtually iden-
rate of delivery should be further investigated for this group. tical after discontinuation of oral opioid therapy (14). Hamza et al.
To date no clinical studies have compared bolus to continuous likewise demonstrated analgesic efficacy in subjects trialed and
infusion, although the authors understand that such an experiment maintained on low-dose IT opioids, with most subjects dramatically
is in progress (282). reducing or eliminating oral opioid use (15). However, it should be
The use of bolus dosing in addition to continuous flow is possible emphasized that studies on microdosing were not controlled or ran-
with commercially available IDD systems. One prospective registry domized. Such findings underscore the importance of RCTs.
of 168 patients suggested that patient-controlled bolus therapy Several studies have examined the impact of pretrial opioid use
122

with concomitant constant infusion resulted in improved patient on postimplant IT analgesia. Kim et al. found that pretrial systemic

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

Table 28. Recommendations Regarding Patient Characteristics Affecting Intrathecal Therapy—Baseline Dose of Opioids, High vs. Low, by the PACC Using
USPSTF Criteria.

Statements Evidence levels Recommendation strength Consensus strength

Pretrial opioid dose does not appear to be predictive of II-3 C Moderate


intrathecal drug delivery outcomes.
Effective sustainable analgesia is achievable with intrathecal drug delivery II-2 B Moderate
without systemic supplementation.

opioid requirement was a poor predictor of IT dose, efficacy, or need psychological assessment as a component of the selection criteria
to change medication at one year postimplant (100). This study did (289). The continuing emphasis is on identification of predictive
find that the trial IT opioid dose was a good predictor of success characteristics. Yet identification of patient states or traits that pre-
with IT therapy. Likewise, Mekhail et al. reported no link between dict outcome is not scientifically valid. A more reliable approach is
systemic opioid requirement and efficacy with IT opioids (108). A to assess for and identify psychological symptoms (e.g., depression)
recent analysis has suggested a significant cost benefit to the elimi- and/or psychiatric diagnoses (e.g., post-traumatic stress disorder),
nation of oral opioid therapy in those transitioning to IDD (103). which could be barriers to a positive outcome.
Taken together, these data suggest that the goal of limiting or As noted already, the approach to a patient with cancer-related
eliminating oral opioid therapy in those transitioning to IDD can be pain should be somewhat more flexible. For patients with signifi-
accomplished in many ways. No large-scale trial has compared pre- cantly compromised life expectancy (Category 1, Table 7), psycho-
implant opioid cessation with postimplant cessation, however, in logical evaluation should be considered optional. We encourage
the two largest studies to date analgesic efficacy was achieved with psychological evaluation for patients in whom the disease process
both methods, albeit at lower doses in the pretrial taper study. There has been arrested but there is a significant probability of reoccur-
appears to be little value in the preimplantation opioid dose as a rence (Category 2, Table 7). Patients whose cancer has been eradi-
predictor of success with IT therapy (100,108). The impact on cated by appropriate therapies and continue to manifest chronic
response to ziconotide based on preimplant opioids has not been pain secondary to medical treatment/anatomical/disease-related
determined. damage, but wherein life expectancy is only minimally compro-
The PACC recommendations regarding patient characteristics that mised, should be considered in the same context as chronic non-
affect IT therapy appear in Table 28. cancer pain patients (Category 3, Table 7).
Consensus Point 24. Psychological assessment, counseling, and
PSYCHOLOGICAL CONSIDERATIONS after care are recommended in appropriate candidates. The use of
an assessment is critical in all noncancer patients receiving an intra-
The general belief that identifying comorbid psychological factors, thecal drug delivery system. An extensive discussion of the proper
which could compromise treatment success, was borrowed from tools and techniques of this screening process is presented in the
neurostimulation practice and guidelines and applied to IT therapy, PACC trialing recommendations (118).
especially in the noncancer pain setting. Nelson et al. in 1996 (287) Consensus Point 25. Psychological screening is not required for
proposed a list of “red flags” to success of treatment that included end-of-life patients, but psychological counseling should be
suicidality, alcohol or drug dependency, unresolved compensation/ considered.
legal issues, severe depression, and so on, which, although not
empirically derived, made sense clinically. This spurred a “rule-out”
approach to the assessment for neuromodulation in general. More EDUCATIONAL REQUIREMENTS FOR
recent guidelines (56) have emphasized the assessment of positive IMPLANTING AND/OR MANAGING IDDS
characteristics such as proper expectation, social support, effective THERAPY
coping skills, and so on, and the importance of using psychological
intervention before and after internalization of an IDD device. Some The extensive scope and breadth of this sixth edition of the PACC
third-party payers mandate this screening process for authorization guidelines is a reflection of the growth of knowledge related to the
of the procedure. safe implementation of implantable IT therapies. In addition to the
Four questions summarize the practical considerations related to rapid growth of the preclinical and clinical science knowledge, there
psychological assessment for IT interventions: 1) Should psychologi- has also been an increase in the number of commercially available
cal evaluation be performed? 2) If so, when is the best time for eval- implantable IDDSs. While all of these devices function by pumping
uation? 3) Who should perform the psychological evaluation? medication from an implantable reservoir to the IT space via an
4) What are the best practice guidelines for psychological evalua- implanted catheter, their propellant mechanism, MRI compatibility,
tion? These and other aspects of psychological screening are dis- and device-specific engineering limitations vary substantially. Rapid-
cussed more thoroughly in a PACC companion article on screening ly advancing clinical and scientific knowledge combined with the
trials for IT therapy (118). variability of pump designs and function make it imperative that
A review of the published IT literature from 1998 through 2010 providers throughout the health care continuum are thoroughly
reveals few psychological evaluations in the studies identified (288). trained and credentialed to provide appropriate and safe care to
Furthermore, there appear to be few, if any, systematic studies with patients implanted with these devices. The specific training recom-
sufficient follow-up to determine the contribution of psychological mendations are largely dependent on the scope of practice of the
123

evaluation to outcome. There has also been criticism of reliance on individual provider, the disease state being treated, and the device

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

being used. Every patient should have a comprehensive pump man- devices to better identify them for patients who are unable to pro-
ager who is accessible throughout the continuum of medical care. vide detailed information regarding their pump. For credentialing
The NANS in partnership with the INS is focused on education and purposes, a minimum of ten interrogations per year for each make is
credentialing as a strategy to continue to improve safety and effica- recommended prior to independently interrogating a pump without
cy of these therapies by promoting improved implementation and the assistance of an industry representative or another supervising
maintenance training and assessment. Credentialing by the hospital provider. These ten interrogations can be performed in one setting
site of service is currently the mechanism of certification, and inher- as part of a hands-on training workshop. The PACC feels that most
ently this is nonuniform. programs do not currently meet these standards.
Consensus Point 27. The PACC recommends each accredited
First Responders
facility have the ability to evaluate an indwelling implanted device,
All health care providers, and especially first responders to disease
including pump and catheter system. We encourage manufacturers
or trauma, as part of their core curriculum and training, should be
and facilities to collaborate on this important issue with a goal of
trained to identify patients with implanted systems. They should be
meeting compliance by the next scheduled PACC in 2019.
aware that there are multiple different types of implanted systems,
including pacemakers, SCS devices and implanted pumps, and
Personnel Who Perform Maintenance and Programming/
should be able to distinguish between them. When an implanted
Reservoir Refill of IDDS
infusion pump is suspected (most often large circular device vs. neu-
Virtually all patients implanted with an IDDS will need pump inter-
rostimulator, pacemaker, or other smaller implanted device), the first
rogation and refill at regular intervals, whether they are inpatients,
responder should have information available to them and be trained
outpatients, or homebound patients. The provider of this service
to contact a pump manager or a representative of the pump
may or may not be the managing physician but should be under
manufacturer.
the supervision of a managing physician. The provider of this service
Suggested Training Milestones is expected to be appropriately trained to perform the following
Suggested training regarding implanted neuromodulation devi- and have a supervising IDDS physician pump manager. In many set-
ces, their indications and the processes for monitoring and refilling tings these tasks are accomplished by licensed nursing infusion
them, should be performed in nursing schools, medical schools and company employees. These service providers should be properly
schools that train allied health professionals. Likewise, this same trained and supervised.
information should be provided in continuing medical education
Programming and Refilling Skill Set
courses for further education of these health care providers. Current-
ly most programs in Europe, Asia, Australia, and the United States • Perform pump interrogation and programming
do not include this information. • Safely perform an aseptic pump refill (preferably with and without
image guidance)
Consensus Point 26. The PACC recommends an overview of • Diagnose and detect a pocket fill and notify a credentialed physi-
intrathecal drug delivery be added to the basic curriculum of physi-
cian pump manager for management
cians in training, nurses, and allied health care professionals. • Identify residual volume discrepancies and notify a credentialed
physician pump manager for management
Pump Interrogator • Be familiar with medication formulations that are appropriate
All patients admitted to a medical facility should have their IDDS,
and inappropriate for intrathecal drug delivery (PACC
medications, and daily dosage of IT medication documented as part
recommendations)
of the medical record. For programmable pumps this often requires • Have supervision by a credentialed physician pump manager
an electronic interrogation of the pump and documentation of the
drug concentrations and delivery modes. Documentation of type Suggested Training Milestones
and manufacturer of pump identification for drug delivery should Training should include the detailed didactic lectures described
be considered the standard of care prior to performing an MRI or above and should also include added lectures on the medication
pursuing elective surgery, and is imperative for all patients admitted choices for IT therapy and pharmacodynamics and pharmacokinetics
to a hospital. The interrogator of the IDDS is expected to be appro- of IT drug delivery. Additional didactic lectures on the indications
priately trained to perform the following: and contraindications for implementing IDDS therapy as well as

evaluating and recognizing serious AEs such as pump pocket fills,
Data gathering and communication skill set
• granuloma formation, and signs and symptoms of medication over/
Interrogate an IDDS without altering programming
• Determine manufacturer, model, and when the device was underdosage should apply. For training purposes, a minimum of 20
implanted supervised pump refills is recommended for initial assessment prior
• Determine the drug(s) and dosage(s) delivered in a day to refills being performed independently. A minimum of ten pump
• Determine the drug refill alarm dates refills per year is suggested to by consensus in order that physician
• Access and interpret alarm logs pump managers maintain clinical competency for each make of
• Contact a physician pump manager device. If these minimums are not achievable for every make, the
pump refill should be directly supervised by a physician pump man-
ager, a credentialed nurse who has met these requirements, or by a
Suggested Training Milestones and Credentialing for Pump
manufacturer representative.
Interrogators
Detailed didactic lectures reviewing all currently available devices Consensus Point 28. The training of all personnel for device eval-
and techniques for pump interrogation are essential. These lectures uation and refilling is an important part of patient care. This training
124

should include the x-ray imaging of all commercially available should be device specific and supervised carefully. The PACC

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

recommends that 20 refills be supervised before independent prac- Consensus Point 29. In order to offer intrathecal therapies
tice is approved. Two or more trained individuals should check all regardless of primary specialty, the physician should be supervised
reprogramming. in a minimum of ten implant and/or explant cases.
Consensus Point 30. In order to maintain skills, the implanter
IDDS Implanting Physician should be involved in five cases more than two years, or should
Intrathecal pump implantation and/or explantation requires basic undergo additional hands-on certified educational training to refresh
surgical skills necessary to perform the procedure safely. Not all skills.
implanters are IDDS managers and not all mangers are IDDS implan-
ters. In those cases, where the manager and implanter are not the Physician Pump Manager
same individual, the two providers should be in close communica- All patients with IT pumps should have a designated physician
tion regarding appropriate planning for the placement of the pump pump manager. This individual will provide the coordination of care
and catheter and to determine the concentrations and types of amongst other providers across the health care spectrum.
medications to be infused by the pump. In addition, prior to implan-
tation, a designated manager must be identified and available to Pump Manager Skill Set
manage the pump immediately after implantation and to monitor • Be a leader who can manage therapy and complications associat-
for adverse side effects of IT medications. In cases of explantation, ed with IDDS
the manager should inform the explanter of the systemic medica- • Diagnose and manage overdose and underdose (withdrawal
tion to be delivered once the IDDS is removed. The IDDS implanter syndrome)
is expected to be appropriately trained to perform the following • Perform dye studies, rotor studies, interpret imaging (fluoroscopy,
procedures and have a supervising IDDS physician pump manager CT, MRI)
available before the case to address potential drug overdose and • Understand the pharmacology of IT drug delivery
underdose. • Responsibilities include, but are not limited to: patient and device
selection, pump fills, prescribing, altering therapy, interactions
Implanter Skill Set with non-IT medications and systems (e.g., MRI)
• Appropriately place a pump subcutaneously and tunnel a catheter
Suggested Training Milestones
from the pump site to the catheter insertion site
If the manager is also the implanter this individual should meet all
• Appropriate and complete medical training in the area of Pain
the requirements previously described, in addition to being compe-
Medicine or Surgery, with a focus on implantable technologies,
tent at performing dye studies/side access port procedures for each
recognized by the country and area of practice.1
make of pumps prior to performing management services indepen-
• Diagnose and troubleshoot potential intraoperative surgical
dently. For specific manufactured devices where fewer than ten
complications
cases have been completed, another manager with skills in that
• Diagnose and troubleshoot potential postoperative and chronic
device should proctor or assistance be obtained from the manufac-
complications of IDDS therapy requiring surgical intervention, for
turer. In addition to the information provided in the didactic lectures
example, granuloma formation, catheter breaks, catheter
previously listed, the manager should also be knowledgeable about
occlusions
all of the advanced intrathecal pharmacokinetic and pharmacody-
namic principles and current medical guidelines. This same knowl-
Suggested Training Milestones
edge base is required for the managers who are not implanters.
The implanter should have also completed the basic didactic
training required of the refill provider as previously described. A Consensus Point 31. The training to manage an intrathecal
minimum of ten supervised implant and/or explant cases should be device is critical to the long-term success of the therapy. All pump
performed under supervision of a credentialed implanter. Another management physicians should have ongoing educational training
five cases over the course of two years should be completed to that includes knowledge of all current and FDA-approved devices
maintain clinical competency. For intracerebroventricular placed and future devices approved by regulatory bodies for research, and
catheters, implanter must be credentialed in neurosurgery. found to be clinically relevant. Managing physicians who are not
implanting physicians are expected to be trained at the same level
1
as those who both implant and manage devices.
Special comment is necessary regarding the suggested formal medical
and surgical education. As this manuscript serves as a living, international Consensus Point 32. To establish a national data base for all
document, it is clear that no uniform credentialing body exists to measure intrathecal pump-managing physicians or healthcare professionals
(or test) specific training criteria over such a diverse group. However, basic as a repository of current pump settings, mediations, efficacy, and
skill standards can be measured. Each implanter must undergo appropri- side effects.
ate surgical tissue management training, with specific experience with
implanting IT therapy. Internationally, the World Institute of Pain (WIP) cre-
ated an exam to standardize internationally delivered interventional pain CONCLUSIONS
management, and there is discussion surrounding this effort through the
educational committee collaborations of NANS, INS, and WIP. In the United The previous PACC work led to improved patient safety and effi-
States, since the inception of an American Council of Graduate Medical Educa-
cacy and advanced questions that fostered additional IT drug
tion (ACGME) certified training program in Pain Medicine and surgical subspe-
cialties of Neurosurgery or Orthopedic Spine Surgery, it is recommended that research. In the same spirit, this present manuscript presents the
implanters have underdone and completed such training. This recommenda- next step in algorithmic thinking. The creation of new algorithmic
tracks for neuropathic and nociceptive pain is an important step in
125

tion, does not, however, impact “legacy or grandfathered” practitioners for


whom no such training was available. improving patient care. The panel encourages continued research

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

and development, including the development of new drugs, devi- 3. Pope JE, Falowski S, Deer TR. Advanced waveforms and frequency with spinal cord
stimulation: burst and high frequency energy delivery. Expert Rev Med Devices
ces, and safety recommendations to improve the care of patients 2015;12:431–437.
whom we strive to help. The PACC is hopeful that the time interval 4. Consent Decree of Permanent Injunction. United States of America vs. Medtronic,
between now and our next update produces new insights in the Inc., and S. Omar Ishrak and Thomas M. Teft. United States District Court for the
District of Minnesota. April 2015.
field of IT drug delivery. 5. Pope J, Poree L, McRoberts WP, Falowski S, Deer T. Consent decree: physician and
The creation of this consensus statement has depended heavily institution ramifications? Neuromodulation 2015;18:653–656.
6. Hassenbusch SJ, Portenoy RK, Cousins M et al. Polyanalgesic Consensus Confer-
on available literature, clinical experience and scientific discourse. ence 2003: an update on the management of pain by intraspinal drug delivery—
Despite our mechanisms used to create the best consensus recom- report of an expert panel. J Pain Symptom Manage 2004;27:540–563.
7. Deer T, Krames ES, Hassenbusch SJ et al. Polyanalgesic Consensus Conference
mendations possible, the final conclusions include a subjective com- 2007: recommendations for the management of pain by intrathecal (intraspinal)
ponent and may be controversial. The panel has addressed drug delivery: report of an interdisciplinary expert panel. Neuromodulation 2007;
nociceptive and neuropathic pain pathways to best treat pain by IT 10:300–328.
8. Deer TR, Prager J, Levy R et al. Polyanalgesic Consensus Conference 2012: recom-
infusion. The panel had considered a third pathway for mixed pain mendations for the management of pain by intrathecal (intraspinal) drug delivery:
syndromes but considering the heterogeneous components of this report of an interdisciplinary expert panel. Neuromodulation 2012;15:436–464; dis-
cussion 464–436.
complex patient group, the reader is advised to use best clinical 9. Hayek SM, Hanes MC. Intrathecal therapy for chronic pain: current trends and
judgment to choose the most appropriate pathway, with the realiza- future needs. Curr Pain Headache Rep 2014;18:388.
tion that the patient may exhibit different components of pain at dif- 10. Deer TR, Pope JE. Factors to consider in the choice of intrathecal drug in the treat-
ment of neuropathic pain. Expert Rev Clin Pharmacol 2015;8:507–510.
fering and various times. 11. Smith TJ, Staats PS, Deer T et al. Randomized clinical trial of an implantable drug
delivery system compared with comprehensive medical management for refracto-
ry cancer pain: impact on pain, drug-related toxicity, and survival. J Clin Oncol
Acknowledgement 2002;20:4040–4049.
12. Hayek SM, Veizi IE, Narouze SN, Mekhail N. Age-dependent intrathecal opioid esca-
lation in chronic noncancer pain patients. Pain Med 2011;12:1179–1189.
The PACC was initiated by INS and funded by unrestrict- 13. Dominguez E, Sahinler B, Bassam D et al. Predictive value of intrathecal narcotic tri-
als for long-term therapy with implantable drug administration systems in chronic
ed educational grants from Medtronic Inc., and Jazz Phar- non-cancer pain patients. Pain Pract 2002;2:315–325.
maceuticals, Inc. No corporate entities had any direct input 14. Grider JS, Harned ME, Etscheidt MA. Patient selection and outcomes using a low-
dose intrathecal opioid trialing method for chronic nonmalignant pain. Pain Physi-
into the contents of the manuscript or the conclusions of cian 2011;14:343–351. 3
the collaborators. Sarah Staples, MA, ELS, assisted with 15. Hamza M, Doleys D, Wells M et al. Prospective study of 3-year follow-up of low-
dose intrathecal opioids in the management of chronic nonmalignant pain. Pain
manuscript preparation. Med 2012;13:1304–1313.
16. Bernards CM. Recent insights into the pharmacokinetics of spinal opioids and the
relevance to opioid selection. Curr Opin Anaesthesiol 2004;17:441–447.
17. Krames ES. Intrathecal infusional therapies for intractable pain: patient manage-
Authorship Statements ment guidelines. J Pain Symptom Manage 1993;8:36–46.
18. Friese S, Hamhaber U, Erb M, Kueker W, Klose U. The influence of pulse and respi-
Dr. Deer served as primary author, project organizer and editor; ration on spinal cerebrospinal fluid pulsation. Investigat Radiol 2004;39:120–130.
19. Henry-Feugeas MC, Idy-Peretti I, Baledent O et al. Origin of subarachnoid cerebro-
Dr. Doleys, Falowski, Jacobs, Kim, and Narouze, performed literature spinal fluid pulsations: a phase-contrast MR analysis. Magn Reson Imaging 2000;18:
searches; Drs. Deer, Hayek, Pope, Grider, and Erdek prepared evi- 387–395.
dence tables; Drs. Huntoon, Mekhail, and Krames served as senior 20. Bernards CM. Cerebrospinal fluid and spinal cord distribution of baclofen and
bupivacaine during slow intrathecal infusion in pigs. Anesthesiology 2006;105:169–
editors; the remaining authors acquired or interpreted data, wrote 178.
sections of the manuscript, and provided critical reviews and 21. Doleys DM. Psychologic evaluation for patients undergoing neuroaugmentative
procedures. Neurosurg Clin North Am 2003;14:409–417.
editing. 22. Doleys DM, Brown J. MMPI profile as an outcome “predictor” in the treatment of
noncancer pain patients utilizing intraspinal opioid therapy. Neuromodulation
2001;4:93–97.
23. Doleys DM, Brown JL, Ness T. Multidimensional outcomes analysis of intrathecal,
How to Cite this Article: oral opioid, and behavioral-functional restoration therapy for failed back surgery
Deer T.R., Pope J.E., Hayek S.M., Bux A., Buchser E., Eldabe syndrome: a retrospective study with 4 years’ follow-up. Neuromodulation 2006;9:
270–283.
S., De Andres J.A., Erdek M., Patin D., Grider J.S., Doleys 24. Product Surveillance Registry (PSR) Database. Minneapolis, MN: Medtronic plc;
D.M., Jacobs M.S., Yaksh T.L., Poree L., Wallace M.S., 2015.
Prager J., Rauck R., DeLeon O., Diwan S., Falowski S.M., 25. Harris RP, Helfand M, Woolf SH et al. for the Methods Work Group, Third US Pre-
ventive Service Task Force. A review of the process. Am J Prev Med 2001;20:21–35.
Gazelka H.M., Kim P., Leong M., Levy R.M., McDowell II G., 26. Hayek SM, Deer TR, Pope JE, Panchal S, Patel V, Burton AW. Intrathecal therapy for
McRoberts P., Naidu R., Narouze S., Perruchoud C., Rosen cancer and non-cancer pain. Pain Physician 2011;14:219–248.
S.M., Rosenberg W.S., Saulino M., Staats P., Stearns L.J., 27. Prager J, Deer T, Levy R et al. Best practices for intrathecal drug delivery for pain.
Neuromodulation 2014;17:354–372; discussion 372.
Willis D., Krames E., Huntoon M., Mekhail N. 2017. The 28. Deer TR, Smith HS, Cousins M et al. Consensus guidelines for the selection and
Polyanalgesic Consensus Conference (PACC): implantation of patients with noncancer pain for intrathecal drug delivery. Pain
Recommendations on Intrathecal Drug Infusion Systems Physician 2010;13:E175–E213.
29. Falco F, Patel V, Hayek S et al. Intrathecal infusion systems for long-term manage-
Best Practices and Guidelines. ment of chronic cancer pain: an update of assessment of evidence. Pain Physician
Neuromodulation 2017; 20: 96–132 2013;16:SE185–SE216.
30. Kloth D. President’s Message. NANS Newsletter 2015;11:1–2.
31. Yaksh TL. New horizons in our understanding of the spinal physiology and phar-
macology of pain processing. Semin Oncol 1993;20(2Suppl.1):6–18.
REFERENCES 32. Rauck RL, Wallace MS, Leong MS et al. A randomized, double-blind, placebo con-
trolled study of intrathecal ziconotide in adults with severe chronic pain. J Pain
Symptom Manage 2006;31:393–406.
1. Pope JE, Deer TR. Intrathecal pharmacology update: novel dosing strategy for 33. Staats PS, Yearwood T, Charapata SG et al. Intrathecal ziconotide in the treatment
intrathecal monotherapy ziconotide on efficacy and sustainability. Neuromodula- of refractory pain in patients with cancer or AIDS: a randomized controlled trial.
tion 2015;18:414–420. JAMA 2004;291:63–70.
2. Bennett G, Burchiel K, Buchser E et al. Clinical guidelines for intraspinal infusion: 34. Wallace MS, Charapata SG, Fisher R et al. Intrathecal ziconotide in the treatment of
126

report of an expert panel. PolyAnalgesic Consensus Conference 2000. J Pain Symp- chronic nonmalignant pain: a randomized, double-blind, placebo-controlled clini-
tom Manage 2000;20:S37–S43. cal trial. Neuromodulation 2006;9:75–86.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

35. Pope JE, Deer TR, McRoberts WP. Intrathecal therapy: the burden of being posi- 67. Alexander JW, Solomkin JS, Edwards MJ. Updated recommendations for control of
tioned as a salvage therapy. Pain Med 2015;16:2036–2038. surgical site infections. Ann Surg 2011;253:1082–1093.
36. Poree L, Krames E, Pope J, Deer TR, Levy R, Schultz L. Spinal cord stimulation as 68. Forse RA, Karam B, MacLean LD, Christou NV. Antibiotic prophylaxis for surgery in
treatment for complex regional pain syndrome should be considered earlier than morbidly obese patients. Surgery 1989;106:750–756; discussion 756–757.
last resort therapy. Neuromodulation 2013;16:125–141. 69. Hetem DJ, Bootsma MC, Bonten MJ. Prevention of surgical site infections:
37. Deer TR, Caraway D, Wallace M. A definition of refractory pain to help determine decontamination with mupirocin based on preoperative screening for Staphy-
suitability for device implantation. Neuromodulation 2014;17:711–715. lococcus aureus carriers or universal decontamination? Clin Infect Dis 2016;62:
38. Pope JE, Deer TR. Intrathecal drug delivery for pain: a clinical guide and future 631–636.
directions. Pain Mange 2015;5:175–183. 70. Kohler P, Sommerstein R, Sch€ onrath F et al. Effect of perioperative mupirocin and
39. Centers for Disease Control (CDC). Vital signs: overdoses of prescription opioid antiseptic body wash on infection rate and causative pathogens in patients under-
pain relievers—United States, 1999-2008. Morb Mortal Wkly Rep 2011;60:1–6. going cardiac surgery. Am J Infect Control 2015;43:e33–e38.
40. Han B, Compton WM, Jones JM et al. Nonmedical prescriptive opioid use and use 71. Lamplot JD, Luther G, Mawdsley EL, Luu HH, Manning D. Modified protocol
disorders among adults aged 18-64 years in the United States 2003-2013. JAMA decreases surgical site infections after total knee arthroplasty. J Knee Surg 2015;28:
2015;314:1468–1478. 395–403. Erratum in: J Knee Surg 2015 Oct;28(5):e1.
41. Coffey RJ, Owens ML, Broste SK et al. Mortality associated with implantation and 72. Anderson MJ, David ML, Scholz M et al. Efficacy of skin and nasal povidone-iodine
management of intrathecal opioid drug infusion systems to treat noncancer pain. preparation against mupirocin-resistant methicillin-resistant Staphylococcus aureus
Anesthesiology 2009;111:881–891. and S. aureus within the anterior nares. Antimicrob Agents Chemother 2015;59:
42. Coffey RJ, Owens ML, Broste SK et al. Medical practice perspective: identification 2765–2773.
and mitigation of risk factors for mortality associated with intrathecal opioids for 73. Bryce E, Wong T, Forrester L et al. Nasal photodisinfection and chlorhexidine wipes
non-cancer pain. Pain Med 2010;11:1001–1009. decrease surgical site infections: a historical control study and propensity analysis.
43. Prager J, Jacobs M. Evaluation of patients for implantable pain modalities: medical J Hosp Infect 2014;88:89–95. doi: 10.1016/j.jhin.2014.06.017. Epub 2014 Aug 1. Erra-
and behavioral assessment. Clin J Pain 2001;17:206–214. tum in: J Hosp Infect 2015;91(1):93.
44. North RB, Kidd DH, Farrokhi F, Piantadosi SA. Spinal cord stimulation versus repeat- 74. Phillips M, Rosenberg A, Shopsin B et al. Preventing surgical site infections: a ran-
ed lumbosacral spine surgery for chronic pain: a randomized, controlled trial. Neu- domized, open-label trial of nasal mupirocin ointment and nasal povidone-iodine
rosurgery 2005;56:98–106; discussion 106–107. Available from: http://www.ncbi. solution. Infect Control Hosp Epidemiol 2014;35:826–832.
nlm.nih.gov/pubmed/15617591. 75. Tanner J, Parkinson H. Double gloving to reduce surgical cross-infection. Cochrane
45. Kumar K, Taylor RS, Jacques L et al. Spinal cord stimulation versus conventional Database Syst Rev 2006;19:CD003087.
medical management for neuropathic pain: a multicentre randomised controlled 76. Excellence NICE. Surgical site infection: prevention and treatment of surgical site infec-
trial in patients with failed back surgery syndrome. Pain 2007;132:179–188. tion. Clinical Guideline 74. Manchester, UK: National Institute for Health and Clinical
46. Kumar K, Hunter G, Demeria D. Spinal cord stimulation in treatment of chronic Excellence, 2008.
benign pain: challenges in treatment planning and present status, a 22-year expe- 77. Darouiche RO, Wall MJ, Itani KM et al. Chlorhexidine-alcohol versus povidone-
rience. Neurosurgery 2006;58:481–496. iodine for surgical-site antisepsis. N Engl J Med 2010;362:18–26.
47. Hayek SM, Veizi HM. Treatment-limiting complications of percutaneous spinal cord 78. Hutchison JJ, McGuckin M. Occlusive dressings: a microbiologic and clinical review.
stimulator implants: a review of eight years of experience from an academic center Am J Infect Control 1990;18:257–268.
database. Neuromodulation 2015;18:603–609. 79. Horlocker TT, Wedel DJ, Rowlingson JC et al. Regional anesthesia in the patient
48. Al-Kaisy A, Van Buyten J-P, Smet I et al. Sustained effectiveness of 10 kHz high- receiving antithrombotic or thrombolytic therapy: American Society of Regional
frequency spinal cord stimulation for patients with chronic, low back pain: 24- Anesthesia and Pain Medicine Evidence-Based Guidelines (Third Edition). Reg
month results of a prospective multicenter study. Pain Med 2014;15:347–354. Anesth Pain Med 2010;35:64–101.
49. deVos CC, Dijkstra C, Lenders MW, Holsheimer J. Spinal cord stimulation with 80. Atchison SR, Durant PA, Yaksh TL. Cardiorespiratory effects and kinetics of intrathe-
hybrid lead relieves pain in low back and legs. Neuromodulation 2012;15:118–123. cally injected D-Ala2-D-Leu5-enkephalin and morphine in unanesthetized dogs.
50. Liem L, Russo M, Huygen FJ et al. One-year outcomes of spinal cord stimulation of Anesthesiology 1986;65:609–616.
the dorsal root ganglion in the treatment of chronic neuropathic pain. Neuromodu- 81. Ummenhofer WC, Arends RH, Shen DD, Bernards CM. Comparative spinal distribu-
lation 2015;18:48–49. tion and clearance kinetics of intrathecally administered morphine, fentanyl, alfen-
51. Rosenberg J, Tavel E, Jackson A et al. Development of new pain affects the long- tanil, and sufentanil. Anesthesiology 2000;92:739–753.
term patient experience with spinal cord stimulation. Presented at the 7th World 82. Ogden CL, Carrol MD, McDowell MA, Flegal KM. Obesity among adults in the Unit-
Congress World Institute of Pain; May 2014; Maastricht, The Netherlands. (WIP- ed States—no statistically significant change since 2003-2004. NCHS Data Brief
0412). 2007;1:1–8.
52. Saulino M, Stearns L, (moderators). Intrathecal Therapies: Basic Challenges. Concur- 83. Farney RJ, Walker JM, Cloward TV, Rhondeau S. Sleep-disordered breathing associ-
rent session presented at: North American Neuromodulation Society 19th Annual ated with long-term opioid therapy. Chest 2003;123:632–639.
Meeting; Dec. 2015; Las Vegas, NV. 84. Horlocker TT, Burton AW, Connis RT et al. Practice guidelines for the prevention,
53. Lindblom U, Meyerson BA. Influence on touch, vibration and cutaneous pain of detection, and management of respiratory depression associated with neuraxial
dorsal column stimulation in man. Pain 1975;1:257–270. opioid administration. American Society of Anesthesiologists Task Force on Neu-
54. Anderson F, Downing GM, Hill J, Casorso L, Lerch N. Palliative performance scale raxial Opioids. Anesthesiology 2009;110:218–230.
(PPS): a new tool. J Palliative. Care 1996;12:5–11. 85. Ornstein E, Mateo RS, Effects of opioids. In: McLeskey CH, ed. Geriatric anesthesiolo-
55. ECOG-ACRIN. ECOG performance status. http://ecog-acrin.org/resources/ecog-per- gy, 1st ed. Philadelphia: Williams & Wilkins, 1997:259.
formance-status. 86. Rauck R, Webster L, Wallace M et al. Effect of concomitant antidepressant and anti-
56. Deer TR, Smith T, Burton AW. Comprehensive consensus based guidelines on convulsant use on adverse events in patients receiving intrathecal ziconotide in a
intrathecal drug delivery systems in the treatment of pain caused by cancer pain. long-term extension study. Poster resented at: West Virginia Society of Interven-
Pain Physician 2011;14:E283–E312. tional Pain Physicians; Puerto Rico, 2014.
57. Codman, a division of Johnson & Johnson. Implantable infusion pumps. http:// 87. Dworzak H, Fuss F, B€ uttner T. Persisting respiratory depression following intrathe-
www.codmanpumps.com/Products_pumps_overview.asp. cal administration of morphine and simultaneous sedation with midazolam. Anaes-
58. Medtronic plc. Targeted drug delivery. Indications, safety and warnings. Synchromed thesist 1999;48:639–641. German.
II. http://professional.medtronic.com/pt/neuro/idd/ind/index.htm#.VorVwVKkx-A. 88. Rawal N, Tandon B. Epidural and intrathecal morphine in intensive care units.
59. Deer TR, Prager J, Levy R et al. Polyanalgesic Consensus Conference–2012: recom- Intensive Care Med 1985;11:129–133.
mendations on trialing for intrathecal (intraspinal) drug delivery: report of an inter- 89. Kotlinska-Lemieszek A, Klepstad P, Haugen DF. Clinically significant drug-drug
disciplinary expert panel. Neuromodulation 2012;15:420–435; discussion 435. interactions involving opioid analgesics used for pain treatment in patients with
60. Deer TR, Prager J, Levy R, et al. Polyanalgesic Consensus Conference–2012: consen- cancer: a systematic review. Drug Des Devel Ther 2015;169:5255–5267.s
sus on diagnosis, detection, and treatment of catheter-tip granulomas (inflamma- 90. Flack SH, Anderson CM, Bernards C. Morphine distribution in the spinal cord after
tory masses). Neuromodulation 2012;15:483–495; discussion 496. chronic infusion in pigs. Anesth Analg 2011;112:460–464.
61. Deer TR, Levy R, Prager J et al. Polyanalgesic Consensus Conference–2012: recom- 91. Flack SH, Bernards CM. Cerebrospinal fluid and spinal cord distribution of hyper-
mendations to reduce morbidity and mortality in intrathecal drug delivery in the baric bupivacaine and baclofen during slow intrathecal infusion in pigs. Anesthesi-
treatment of chronic pain. Neuromodulation 2012;15:467–482; discussion 482. ology 2010;112:165–173.
62. Ver Donck A, Vranken JH, Puylaert M et al. Intrathecal drug administration in 92. Yaksh TL, de Kater A, Dean R, Best BM, Miljanich GP. Pharmacokinetic analysis of
chronic pain syndromes. Pain Pract 2014;14:461–476. ziconotide (SNX-111), an intrathecal N-type calcium channel blocking analgesic,
63. Webster LR, Choi Y, Desai H, Webster L, Grant BJ. Sleep-disordered breathing and delivered by bolus and infusion in the dog. Neuromodulation 2012;15:508–519; dis-
chronic opioid therapy. Pain Med 2008;9:425–432. cussion 519.
64. Provenzano DA, Deer T, Luginbuhl Phelps A et al. An international survey to under- 93. Kabulski J, Poree L, Wolbers L, Quantitative determination & pharmacokinetic
stand infection control practices for spinal cord stimulation. Neuromodulation modeling of intrathecally administered opioids in plasma. Poster presented at:
2016;19:71–84. International Neuromodulation Society; June 2015: Berlin, Germany.
65. Deer TR, Mekhail N, Provenzano D et al. The appropriate use of neurostimulation: 94. Fitzgibbon DR, Rathmell JP, Michna E, Stephens LS, Posner KL, Domino KB. Mal-
avoidance and treatment of complications of neurostimulation therapies for the practice claims associated with medication management for chronic pain. Anesthe-
treatment of chronic pain. Neuromodulation 2014;17:571–597. siology 2010;112:948–956.
66. Bowater RJ, Stirling SA, Lilford RJ. Is antibiotic prophylaxis in surgery a generally 95. Dunn KM, Saunders KW, Rutter CM. Overdose and prescribed opioids:
127

effective intervention? Testinga generic hypothesis over a set of meta-analyses. associations among chronic noncancer pain patients. Ann Intern Med 2010;152:
Ann Surg 2009;249:551–556. 85–92.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

96. Chou R, Turner JA, Devine EB. The effectiveness and risks of long-term opioid ther- 127. Fukushima S, Takenami T, Yagishita S, Nara Y, Hoka S, Okamoto H. Neurotoxicity of
apy for chronic pain: a systemic review for a National Institutes of Health Pathways intrathecally administered fentanyl in a rat spinal model. Pain Med 2011;12:717–
to Prevention Workshop. Ann Intern Med 2015;162:276–286. 725.
97. Partnership Healthplan of California. http://www.partnershiphp.org/Pages/PHC.aspx. 128. Abut YC, Turkmen AZ, Midi A, Eren B, Yener N, Nurten A. Neurotoxic effects of lev-
98. Tyler CB, Advokat C. Investigation of “cross-tolerance” between systemic and intra- obupivacaie and fentanyl on rat spinal cord. Rev Bras Anestesiol 2015;65:27–33.
thecal morphine in rats. Physiol Behav 1986;37:27–32. 129. Yaksh TL, Noueihed RY, Durant PAC. Studies of the pharmacology and pathology
99. Veizi IE, Hayek SM, Narouze S, Pope JE, Mekhail N. Combination of intrathecal of intrathecally administered 4-anilinopiperidine analogues and morphine in rat
opioids with bupivacaine attenuates opioid dose escalation in chronic noncancer and cat. Anesthesiology 1986;64:54–66.
pain patients. Pain Med 2011;12:1481–1489. 130. Sabbe MB, Grafe MR, Mjanger E, Tiseo PJ, Hill HF, Yaksh TL. Spinal delivery of
100. Kim D, Sidov A, Mandhare V, Shuster A. Role of pretrial systemic opioid require- sufentanil, alfentanil and morphine in dogs. Physiol Toxicol Investig Anesthesiology
ments intrathecal trial dose and non-psychological factors as predictors of out- 1994;81:899–920.
come of intrathecal pump therapy: one clinician’s experience with lumbar 131. Allen JW, Horais KA, Tozier NA, Yaksh TL. Opiate pharmacology of intrathecal gran-
postlaminectomy pain. Neuromodulation 2011;14:165–175. ulomas. Anesthesiology 2006;105:590–598.
101. Grider JS, Etscheidt MA, Harned ME et al. Trialing and maintenance dosing using a 132. Yaksh TL, Steinauer JJ, Veesart SL, Malkmus SA. Alfentanil: correlations between
low-dose intrathecal opioid method for chronic nonmalignant pain: a prospective absence of effect upon subcutaneous mast cells and absence of granuloma forma-
36-month study. Neuromodulation 2015;19:206–219. tion after intrathecal infusion in the dog. Neuromodulation 2013;16:459–466; dis-
102. Centers for Disease Control. Guideline for Prescribing Opioids for Chronic Pain. http:// cussion 466.
www.cdc.gov/drugoverdose/pdf/guidelines_factsheet-a.pdf. 133. Rawal N, Nuutinen L, Raj PP et al. Behavioral and histopathologic effects following
103. Hatheway JA, Caraway D, David G et al. Systemic opioid elimination after implanta- intrathecal administration of butorphanol, sufentanil, and nalbuphine in sheep.
tion of an intrathecal drug delivery system significantly reduced health-care Anesthesiology 1991;75:1025–1123.
expenditures. Neuromodulation 2015;18:207–213; discussion 213. 134. Deer TR, Caraway DL, Kim CK, Dempsey CD, Stewart CD, McNeil KF. Clinical experi-
104. Yaksh TL, Onofrio BM. Retrospective consideration of the doses of morphine given ence with intrathecal bupivacaine in combination with opioid for the treatment of
intrathecally by chronic infusion in 163 patients by 19 physicians. Pain 1987;31: chronic pain related to failed back surgery syndrome and metastatic cancer pain
211–223. of the spine. Spine J 2002;2:274–278.
105. Onofrio BM, Yaksh TL. Long-term pain relief produced by intrathecal morphine 135. Hayek SM, Veizi E, Hanes M. Intrathecal hydromorphone and bupivacaine combi-
infusion in 53 patients. J Neurosurg 1990;72:200–209. nation therapy for post-laminectomy syndrome optimized with patient-activated
106. Gestin Y, Vainio A, Pegurier AM. Long-term intrathecal infusion of morphine in bolus device. Pain Med 2015; doi http://dx.doi.org/10.1093/pm/pnv021 pnv021.
the home care of patients with advanced cancer. Acta Anaesthesiol Scand 136. Bernards CM, Hill HF. Physical and chemical properties of drug molecules gov-
1997;41:12–17. erning their diffusion through the spinal meninges. Anesthesiology 1992;77:
107. Roberts LJ, Finch PM, Goucke CR, Price LM. Outcome of intrathecal opioids in 750–756.
chronic non-cancer pain. Eur J Pain 2001;5:353–361. 137. Reina MA, Pulido P, L opez A. Human dural sac [English abstr]. Rev Arg Anestesiol
108. Mekhail N, Mahboobi R, Farajzadeh Deroee A et al. Factors that might impact intra- 2007;65:167–184.
thecal drug delivery (IDD) dose escalation: a longitudinal study. Pain Pract 2014;14: 138. Reina MA, L opez A, De Andr es JA. Variation of human dura mater thickness
301–308. [English abstr]. Rev Esp Anestesiol Reanim 1999;46:344–349.
109. Maves TJ, Gebhart GF. Antinociceptive synergy between intrathecal morphine and 139. Reina MA, L opez A, Dittmann M, De Andr es JA. Structure of human dura mater
lidocaine during visceral and somatic nociception in the rat. Anesthesiology 1992; thickness by scanning electron microscopy [English abstr]. Rev Esp Anestesiol
76:91–99.
Reanim 1996;43:135–137.
110. Penning JP, Yaksh TL. Interaction of intrathecal morphine with bupivacaine and
140. Reina MA, Dittmann M, L opez A, van Zundert A. New perspectives in the micro-
lidocaine in the rat. Anesthesiology 1992;77:1186–2000.
scopic structure of human dura mater in the dorso lumbar region. Reg Anesth
111. Prialt (ziconotide) Intrathecal Infusion [package insert]. http://www.prialt.com/.
1997;22:161–166.
112. Yaksh TL, Fisher C, Hochman T, Wiese A. Current and future issues in the develop-
141. Dittmann M, Reina MA, L opez A. Neue ergebnisse bei der darstellung der dura
ment of spinal agents for the management of pain. Curr Neuropharmacol 2016; in
mater spinalis mittles rasterelektronenmikroskopie [English abstr]. Anaesthesist
press.
1998;47:409–413.
113. Wallace MS, Rauck RL, Deer T. Ziconotide combination intrathecal therapy: ratio-
142. Reina MA, L opez A, Dittmann M, De Andr es JA. External and internal surface of
nale and evidence. Clin J Pain 2010;26:635–644.
human dura mater by scanning electron microscopy [English abstr]. Rev Esp Anes-
114. de la Calle Gil AB, Pe~na Vergara I, Cormane Bornacelly MA, Pajuelo Gallego A. Intra-
tesiol Reanim 1996;43:130–134.
thecal ziconotide and morphine for pain relief: a case series of eight patients with
143. Reina MA, Prats-Galino A, Sola RG, Puigdellıvol-Sanchez A, Arriazu Navarro R, De
refractory cancer pain, including five cases of neuropathic pain. Neurol Ther 2015;4:
159–168. Andr es JA. Structure of the arachnoid layer of the human spinal meninges: a barri-
115. Shields DE, Aclan J, Szatkowski A. Chemical stability of admixtures combining zico- er that regulates dural sac permeability [English abstr]. Rev Esp Anestesiol Reanim
notide with fentanyl or sufentanil during simulated intrathecal administration. Int J 2010;57:486–492.
Pharm Compounding 2008;12:463–466. 144. Reina MA, Villanueva MC, L opez A. Human trabecular arachnoids, pia mater and
116. Mohammed SI, Eldabe S, Simpson KH et al. Bolus intrathecal injection of zicono- spinal anesthesia [English abstr]. Rev Arg Anestesiol 2008;66:111–133.
R ) to evaluate the option of continuous administration via an implanted
tide (PrialtV 145. Reina MA, L opez A, De Andr es JA. Hypothesis on the anatomical bases of cauda
intrathecal drug delivery (ITDD) system: a pilot study. Neuromodulation 2013;16: equine syndrome and transitory radicular irritation syndrome post spinal anesthe-
576–581; discussion 582. sia [English abstr]. Rev Esp Anestesiol Reanim 1999;46:99–105.
117. Compton WM, Jones CM, Baldwin GT. Relationship between nonmedical 146. Reina MA, Mach es F, L
opez A, De Andr es JA. The ultrastructure of the spinal arach-
prescription-opioid use and heroin use. New Engl J Med 2016;174:154–163. noid in humans and its impact on spinal anesthesia, cauda equina syndrome and
118. Deer TR, Hayek SM et al. The Polyanalgesic Consensus Conference (PACC): recom- transient neurological syndrome. Tech Reg Anesth Pain Manage 2008;12:153–160.
mendations for trialing of intrathecal drug delivery infusion therapy. Neuromodula- 147. Kershner DE, Binhammer RT. Lumbar Intrathecal ligaments. Clin Anat 2002;15:82–
tion 2017;20:133–154. 87.
119. Sultan P, Gutierrez MC, Carvalho B. Neuraxial morphine and respiratory depression: 148. Di Chiro G, Timins EL. Supine myelography and the septum posticum. Radiology
finding the right balance. Drugs 2011;71:1807–1819. 1974;111:319–327.
120. Beatty NC, Arendt KW, Niesen AD et al. Analgesia after cesarean delivery: a retro- 149. Reina MA, De Leon Casasola O, Villanueva MC, L opez A, Maches F, De Andr es JA.
spective comparison of intrathecal hydromorphone and morphine. J Clin Anaesth Ultrastructural findings in human spinal pia mater in relation to subarachnoid
2013;25:379–383. anesthesia. Anesth Analg 2004;98:1479–1485.
121. Shapiro A, Zohar E, Zaslansky R, Hoppenstein D, Shabat S, Fredman B. The frequen- 150. Reina MA, L opez Garcıa A, de Andr es JA. Anatomical description of a natural perfo-
cy and timing of respiratory depression in 1524 postoperative patients treated ration present in the human lumbar pia mater [Spanish]. Rev Esp Anestesiol Reanim
with systemic or neuraxial morphine. J Clin Anesth 2005;17:537–542. 1998;45:4–7.
122. Cowan CM, Kendall JB, Barclay PM et al. Comparison of intrathecal fentanyl and 151. Merchant RE, Low FN. Scanning electron microscopy of the subarachnoid space in
diamorphine in addition to bupivacaine for Caesarean section under spinal analge- the dog: evidence for a non-hematogeneous origin of the subarachnoid macro-
sia. Br J Anaesth 2002;89:452–458. phages. Am J Anat 1979;156:183–206.
123. Duman A, Apiliogullari S, Balasar M, Gurbuz R, Karcioglu M. Comparison of 50 152. Bernards CM. Epidural and intrathecal drug movement. In: Yaksh TN, ed. Spinal
microg and 25 microg doses of intrathecal morphine on postoperative analgesic drug delivery. Amsterdam: Elsevier; 1999:239–269.
requirements in patients undergoing transurethral resection of the prostate with 153. Artru AA, Spinal cerebrospinal fluid chemistry and physiology. In: Yaksh TN, ed. Spi-
intrathecal anesthesia. J Clin Anesth 2010;22:329–333. nal drug delivery. Amsterdam: Elsevier; 1999:177–237.
124. Burton AW, Deer TR, Wallace MS, Rauck RL, Grigsby E. Considerations and method- 154. Quencer RM, Post MJD, Hinks RS. Cine MR in the evaluation of normal and
ology for trialing ziconotide. Pain Physician 2010;13:23–33. abnormal CSF flow. Intracranial and intraspinal studies. Neuroradiology 1990;
125. Bolash R, Mekhail M, Multi-center prospective analysis of on-demand intrathecal 32:371–391.
morphine bolus dosing among patients with targeted drug delivery systems. Post- 155. Wagshul ME, Chen JJ, Egnor MR, McCormack EJ, Roche PE. Amplitude and phase
er presented at the North American Neuromodulation Society meeting; Dec. 2015; of cerebrospinal fluid pulsations: experimental studies and review of the literature.
Las Vegas, NV. J Neurosurg 2006;104:810–819.
126. Brogan SE, Winter NB, Okifuji A. Prospective observational study of patient– 156. Hsu Y, Hettiarachchi HD, Zhu DC, Linninger AA. The frequency and magnitude of
128

controlled intrathecal analgesia. Impact on cancer-associated symptoms, break- cerebrospinal fluid pulsations influence intrathecal drug distribution: key factors
through pain control and patient satisfaction. Reg Anes Pain Med 2015;40:369–375. for interpatient variability. Anesth Analg 2012;115:386–394.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

157. Hogan QH, Prost R, Kulier A et al. Magnetic resonance imaging of cerebrospinal 188. Medtronic. Intrathecal distal end catheter occlusions as a result of pH & salt con-
fluid volume and the influence of body habitus and abdominal pressure. Anesthesi- centration gradients between delivery solution & cerebrospinal fluid (CSF). Minne-
ology 1996;84:1341–1349. apolis, MN: Medtronic plc; 2009.
158. Higuchi H, Hirata J, Adachi Y, Kazama T. Influence of lumbosacral cerebrospinal flu- 189. Inturrisi CE. Clinical pharmacology of opioids for pain. Clin J Pain 2002;18(4 Suppl):
id density, velocity, volume and on extent and duration of plain bupivacaine spinal S3–13.
anesthesia. Anesthesiology 2004;100:106–114. 190. Yaksh TL, Allen JW, Veesart SL et al. Role of meningeal mast cells in intrathe-
159. Sullivan JT, Grouper S, Walker MT, Parrish TB, McCarthy RJ, Wong CA. Lumbosacral cal morphine-evoked granuloma formation. Anesthesiology 2013;118:
cerebrospinal fluid volume in humans using three-dimensional magnetic reso- 664–678.
nance imaging. Anesth Analg 2006;103:1306–1310. 191. Yaksh TL, Horais KA, Tozier NA et al. Chronically infused intrathecal morphine in
160. Edsbagge M, Starck G, Zetterberg H, Ziegelitz D, Wikkelso C. Spinal CSF volume in dogs. Anesthesiology 2003;99:174–187.
healthy elderly individuals. Clin Anat 2011;24:733–740. 192. Michael A, Buffen E, Rauck R, Anderson W, McGirt M, Mendenhall HV. An in vivo
161. Puigdellıvol-Sanchez A, Prats-Galino A, Reina MA et al. Tridimensional magnetic canine study to assess granulomatous responses in the MedStream Programmable
resonance image of structures enclosed in the spinal canal relevant to anesthetists Infusion System (TM) and the SynchroMed II Infusion SystemV R . Pain Med 2012;13:
and estimation of the lumbosacral CSF volume. Acta Anaesth Belg 2011;62:37–45. 175–184.
162. Prats-Galino A, Reina MA, Puigdellıvol-Sanchez A, Juanes M endez JA, De Andr es 193. Gradert TL, Baze WB, Satterfield WC, Hildebrand KR, Johansen MJ, Hassenbusch SJ.
JA, Collier CB. Cerebrospinal fluid volume and nerve root vulnerability during lum- Safety of chronic intrathecal morphine infusion in a sheep model. Anesthesiology
bar puncture or spinal anaesthesia at different vertebral levels. Anaesth Intensive 2003;99:188–198.
Care 2012;40:643–647. 194. Yaksh TL. Spinal delivery and assessment of drug safety. In: Bolon B, Butt MT, eds.
163. Prats-Galino A, Reina MA, Mavar Haramija M, Puigdellivol-Sanchez A, Juanes Fundamental neuropathology for pathologists and toxicologists, Chapter 27. Philadel-
Mendez JA, De Andres JA. 3D interactive model of lumbar spinal structures of phia: John Wiley and Sons; 2011:451–462.
anesthetic interest. Clin Anat 2015;28:205–212. 195. Atli A, Theodore BR, Turk DC, Loeser JD. Intrathecal opioid therapy for chronic non-
164. Papisov MI, Belov VV, Gannon KS. Physiology of the intrathecal bolus: the leptome- malignant pain: a retrospective cohort study with 3-year follow-up. Pain Med 2010;
ningeal route for macromolecule and particle delivery to CNS. Mol Pharm 2013;10: 11:1010–1016.
1522–1532. 196. Berg A, Barsa J, Deer T et al. Efficacy of morphine sulfate infusion via the Prom-
165. Dreha-Kulaczewski S, Joseph AA, Merboldt KD, Ludwig HC, Gartner J, Frahm J. etraVR Intrathecal Infusion Pump. A prospective multicenter evaluation: PB238
Inspiration is the major regulator of human CSF flow. J Neurosci 2015;35:2485– [abstract]. Pain Pract 2009;9(Suppl.1):159.
2491. 197. Kongkam P, Wagner DL, Sherman S et al. Intrathecal narcotic infusion pumps for
166. Brinker T, Stopa E, Morrison J, Klinge P. A new look at cerebrospinal fluid circula- intractable pain of chronic pancreatitis: a pilot series. Am J Gastroenterol 2009;104:
tion. Fluids Barriers CNS 2014;11:10. 1249–1255.
167. Kuttler A, Dimke T, Kern S, Helmlinger G et al. Understanding pharmacokinetics 198. Shaladi A, Saltari MR, Piva B et al. Continuous intrathecal morphine infusion in
using realistic computational models of fluid dynamics: biosimulation of drug dis- patients with vertebral fractures due to osteoporosis. Clin J Pain 2007;23:511–
tribution within the CSF space for intrathecal drugs. J Pharmacokinet Pharmacodyn 517.
2010;37:629–664. 199. Saltari MR, Shaladi A, Piva B et al. The management of pain from collapse of osteo-
168. Nelissen RM. Fluid mechanics of intrathecal drug delivery [thesis]. Lausanne: Federal porotic vertebrae with continuous intrathecal morphine infusion. Neuromodulation
Institute of Technology, 2008. 2007;10:167–176.
169. Henry-Feugeas MC, Idy-Peretti L, Blanchet B et al. Temporal and spatial assessment
200. Reig E, Abejon D. Continuous morphine infusion: a retrospective study of efficacy,
of normal cerebrospinal fluid dynamics with MR imaging. Magn Reson Imaging
safety, and demographic variables. Neuromodulation 2009;12:122–129.
1993;11:1107–1118.
201. Raphael JH, Duarte RV, Southall JL, Nightingale P, Kitas GE. Randomised, double-
170. Wallace M, Yaksh TL. Characteristics of distribution of morphine and metabolites
blind controlled trial by dose reduction of implanted intrathecal morphine delivery
in cerebrospinal fluid and plasma with chronic intrathecal morphine infusion in
in chronic non-cancer pain. BMJ Open 2013;3:e003061.
humans. Anesth Analg 2012;114:797–804.
202. Kumar P, Sunkaraneni S, Sirohi S, Dighe SV, Walker EA, Yoburn BC. Hydromor-
171. Buchser E, Durrer A, Chedel E et al. Efficacy of intrathecal bupivacaine: how impor-
phone efficacy and treatment protocol impact on tolerance and mu-opioid recep-
tant is the flow rate? Pain Med 2004;5:248–252.
tor regulation. Eur J Pharmacol 2008;597:39–45.
172. Mironer E, Haasis J, Chapple I et al. Efficacy and safety of intrathecal opioid/bupiva-
203. Johansen MJ, Satterfield WC, Baze WB, Hildebrand KR, Gradert TL, Hassenbusch SJ.
caine mixture in chronic nonmalignant pain: a double blind, randomized, cross-
Continuous intrathecal infusion of hydromorphone: safety in the sheep model and
over, multicenter study by the National Forum of Independent Pain Clinicians
(NFIPC). Neuromodulation 2002;5:208–213. clinical implications. Pain Med 2004;5:14–25.
173. Bernards CM, Shen DD, Sterling ES et al. Epidural, cerebrospinal fluid, and plasma 204. Hoederath P, Gautschi OP, Land M, Hildebrandt G, Fournier JY. Formation of two
pharmacokinetics of epidural opioids (part 1): differences among opioids. Anesthe- consecutive intrathecal catheter tip granulomas within nine months. Cen Eur Neu-
siology 2003;99:455–465. rosurg 2010;71:39–42.
174. Dahm PO, Nitescu PV, Appelgren LK, Curelaru ID. Intrathecal infusion of bupiva- 205. Ramsey CN, Owen RD, Witt WO, Grider JS. Intrathecal granuloma in a patient
caine with or without buprenorphine relieved intractable pain in three patients receiving high dose hydromorphone. Pain Physician 2008;11:369–373.
with vertebral compression fractures caused by osteoporosis. Reg Anest Pain Med 206. Ruan X, Liu H, Couch JP, Wang F, Chiravuri S. Recurrent cellulitis associated with
1999;24:352–357. long-term intrathecal opioid infusion therapy: a case report and review of the liter-
175. Wall PD, Shortland P. Long-range afferents in the rat spinal cord. 1. Numbers, ature. Pain Med 2010;11:972–976.
distances and conduction velocities. Phil Trans R Soc London B Biol Sci 1991; 207. Sirohi S, Dighe SV, Walker EA, Yoburn BC. The analgesic efficacy of fentanyl: rela-
334:85–93. tionship to tolerance and mu-opioid receptor regulation. Pharmacol Biochem
176. Shortland P, Wall PD. Long-range afferents in the rat spinal cord. II. Arborizations Behav 2008;91:115–120.
that penetrate grey matter. Phil Trans R Soc London B Biol Sci 1992;337:445–455. 208. Medel R, Pouratian N, Elias WJ. Catheter-tip mass mimicking a spinal epidural
177. Dahm P, Nitescu P, Appelgren L, Curelaru I. Efficacy and technical complications of hematoma. J Neurosurg Spine 2010;12:66–71.
long-term continuous intraspinal infusions of opioid and/or bupivacaine in refrac- 209. Colpaert FC, Leysen JE, Michiels M, van den Hoogen RH. Epidural and intravenous
tory nonmalignant pain: a comparison between the epidural and the intrathecal sufentanil in the rat: analgesia, opiate receptor binding, and drug concentrations
approach with externalized or implanted catheters and infusion pumps. Clin J Pain in plasma and brain. Anesthesiology 1986;65:41–49.
1998;14:4–16. 210. Gupta A, Martindale T, Christo PJ. Intrathecal catheter granuloma associated with
178. Yaksh TL, Woller SA, Ramachandran R, Sorkin LS. The search for novel analgesics: continuous sufentanil infusion. Pain Med 2010;11:847–852.
targets and mechanisms. F1000 Prime Rep 2015;26:7–56. 211. Reich DL, Silvay G. Ketamine: an update on the first twenty-five years of clinical
179. Kroin JS. Intrathecal drug administration. Present use and future trends. Clin Phar- experience. Can J Anaesth 1989;36:186–197.
macokin 1992;22:319–326. 212. Yaksh TL, Tozier N, Horais KA et al. Toxicology profile of N-methyl-D-aspartate
180. Rennels ML, Blaumanis OR, Grady PA. Rapid solute transport throughout the brain antagonists delivered by intrathecal infusion in the canine model. Anesthesiology
via paravascular fluid pathways. Adv Neurol 1990;52:431–439. 2008;108:938–949.
181. Bernards CM. Understanding the physiology and pharmacology of epidural and 213. Walker SM, Westin BD, Deumens R, Grafe M, Yaksh TL. Effects of intrathecal keta-
intrathecal opioids. Best Pract Res Clin Anaesthesiol 2002;16:489–505. mine in the neonatal rat: evaluation of apoptosis and long-term functional out-
182. Nordberg G. Pharmacokinetic aspects of spinal morphine analgesia. Acta Anaesth come. Anesthesiology 2010;113:147–159.
Scand (Suppl) 1984;79:1–38. 214. Wilson JA, Nimmo AF, Fleetwood-Walker SM, Colvin LA. A randomised double
183. Gudeman J, Jozwiakowski M, Chollet J, Randell M. Potential risks of pharmacy blind trial of the effect of pre-emptive epidural ketamine on persistent pain after
compounding. Drugs R D 2013;13:1–8. lower limb amputation. Pain 2008;135:108–118.
184. Ghafoor VL, Epshteyn M, Carlson GH, Terhaar DM et al. Intrathecal drug therapy 215. Sato C, Okabe T, Nakanishi K, Sakamoto A. A case of cancer pain management by
for long-term patient management. Am J Heath Syst Pharm 2007;64:2447–2461. long-term intrathecal PCA. J Nippon Med School 2010;77:333–337.
185. Rezai A, Kloth D, Hansen H, Schultz D, Thompson S, Deer TR. Physician response to 216. Karpinski N, Dunn J, Hansen L, Masliah E. Subpial vacuolar myelopathy after intra-
medtronic’s position on the use of off-label medications in the synchromed pump. thecal ketamine: report of a case. Pain 1997;73:103–105.
Pain Phys 2013;16:415–417. 217. Rizzo CC, Luretti GR, Mattos AL. The combination of methadone and dexametha-
186. Medtronic product bulletin: summary of approved drugs Medtronic’s SynchroMed II sone for cancer pain. Eur J Pain 2009;13(Suppl. 1):S258.
Infusion System. Minneapolis, MN: Medtronic plc, 2012. 218. Mironer YE, Tollison CD. Methadone in the intrathecal treatment of chronic non-
129

187. Kim P, Case series of distal catheter obstruction. Presented at: NANS Annual Meet- malignant pain resistant to other neuroaxial agents: the first experience. Neuromo-
ing; 2011; Las Vegas, NV. dulation 2001;4:25–31.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

219. Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The physiology, pathology, and 251. Munro G, Ahring PK, Mirza NR. Developing analgesics by enhancing spinal inhibi-
pharmacology of voltage-gated calcium channels and their future therapeutic tion after injury: GABA-A receptor subtypes as novel targets. Trends Pharmacol Sci
potential. Pharmacol Rev 2015;67:821–870. 2009;30:453–459.
220. Takasusuki T, Yaksh TL. Regulation of spinal substance p release by intrathecal cal- 252. Zeilhofer HU, Witschi R, H€ osl K. Subtype-selective GABAA receptor mimetics—
cium channel blockade. Anesthesiology 2011;115:153–164. novel antihyperalgesic agents? J Mol Med 2009;87:465–469.
221. Skov MJ, Beck JC, de Kater AW, Shopp GM. Nonclinical safety of ziconotide: 253. Chiari A, Yaksh TL, Myers RR et al. Preclinical toxicity screening of intrathecal aden-
an intrathecal analgesic of a new pharmaceutical class. Int J Toxicol 2007;26: osine in rats and dogs. Anesthesiology 1999;91:824–832.
411–421. 254. Sabbe MB, Grafe MR, Pfeifer BL, Mirzai TH, Yaksh TL. Toxicology of baclofen contin-
222. Rainov NG, Heidecke V, Burkert W. Long-term intrathecal infusion of drug combi- uously infused into the spinal intrathecal space of the dog. Neurotoxicology 1993;
nations for chronic back and leg pain. J Pain Symptom Manage 2001;22:862–871. 14:397–410.
223. Butterworth JF, Strichartz GR. Molecular mechanisms of local anesthesia: a review. 255. Koulousakis A, Kuchta J. Intrathecal antispastic drug application with implantable
Anesthesiology 1990;72:711–734. pumps: results of a 10 year follow-up study. Acta Neurochir Suppl 2007;97 (Pt. 1):
224. Covino BG. Pharmacology of local anaesthetic agents. Br J Anaesth 1986;58:701–716. 181–184.
225. Scott DB, McClure JH, Giasi RM, Seo J, Covino BG. Effects of concentration of local 256. Deer TR, Raso LJ, Garten TG. Inflammatory mass of an intrathecal catheter in
anaesthetic drugs in extradural block. Br J Anaesth 1980;52:1033–1037. patients receiving baclofen as a sole agent: a report of two cases and a
226. Cohen SE, Yeh JY, Riley ET, Vogel TM. Walking with labor epidural analgesia: the review of the identification and treatment of the complication. Pain Med 2007;
impact of bupivacaine concentration and a lidocaine-epinephrine test dose. Anes- 8:259–262.
thesiology 2000;92:387–392. 257. Deer TR, Raso LJ, Coffey RJ, Allen JW. Intrathecal baclofen and catheter tip inflam-
227. Abrahams M, Higgins P, Whyte P, Breen P, Muttu S, Gardiner J. Intact propriocep- matory mass lesions (granulomas): a reevaluation of case reports and imaging
tion and control of labour pain during epidural analgesia. Acta Anaesthesiol Scand findings in light of experimental, clinicopathological, and radiological evidence.
1999;43:46–50. Pain Med 2008;9:391–395.
228. Dahm P, Nitescu P, Appelgren L, Curelaru I. High thoracic/low cervical, long-term intra- 258. van der Plas AA, Marinus J, Eldabe S, Buchser E, van Hilten JJ. The lack of effi-
thecal (i.t.) infusion of bupivacaine alleviates “refractory” pain in patients with unstable cacy of different infusion rates of intrathecal baclofen in complex regional
angina pectoris. Report of 2 cases. Acta Anaesthesiol Scand 1998;42:1010–1017. pain syndrome: a randomized, double-blind, crossover study. Pain Med 2011;
229. Dahm PO, Nitescu PV, Appelgren LK, Curelaru II. Long-term intrathecal (i.t.) infu- 12:459–465.
sion of bupivacaine relieved intractable pain and spasticity in a patient with multi- 259. Saulino M, Burton AW, Danyo DA, Frost S, Glanzer J, Solanki DR. Intrathecal zicono-
ple sclerosis. Eur J Pain 1998;2:81–85. tide and baclofen provide pain relief in seven patients with neuropathic pain and
230. Lundborg C, Dahm P, Nitescu P, Appelgren L, Curelaru I. Clinical experience using spasticity: case reports. Eur J Phys Rehabil Med 2009;45:61–67.
intrathecal (IT) bupivacaine infusion in three patients with complex regional pain 260. Lind G, Schechtmann G, Winter J, Meyerson BA, Linderoth B. Baclofen-enhanced
syndrome type I (CRPS-I). Acta Anaesthesiol Scand 1999;43:667–678. spinal cord stimulation and intrathecal baclofen alone for neuropathic pain: long-
231. Akerman B, Arwestr€ om E, Post C. Local anesthetics potentiate spinal morphine term outcome of a pilot study. Eur J Pain 2008;12:132–136.
antinociception. Anesth Analg 1988;67:943–948. 261. Heetla HW, Staal MJ, Kliphuis C, van Laar T. The incidence and management of tol-
232. Li DF, Bahar M, Cole G, Rosen M. Neurological toxicity of the subarachnoid infusion erance in intrathecal baclofen therapy. Spinal Cord 2009;47:751–756.
of bupivacaine, lignocaine or 2-chloroprocaine in the rat. BrJ Anaesth 1985;57: 262. Dones I, Broggi G. A case of very long-term appearing drug tolerance to intrathe-
424–429. cal baclofen. J Neurosurg Sci 2010;54:77–78.
233. Kroin JS, McCarthy RJ, Penn RD, Kerns JM, Ivankovich AD. The effect of chronic 263. Hansen CR, Gooch JL, Such-Neibar T. Prolonged, severe intrathecal baclofen
subarachnoid bupivacaine infusion in dogs. Anesthesiology 1987;66:737–742. withdrawal syndrome: a case report. Arch Phys Med Rehabil 2007;88:
234. Wagemans MF, van der Valk P, Spoelder EM, Zuurmond WW, de Lange JJ. Neuro- 1468–1471.
histopathological findings after continuous intrathecal administration of morphine 264. Salazar ML, Eiland LS. Intrathecal baclofen withdrawal resembling serotonin
or a morphine/bupivacaine mixture in cancer pain patients. Acta Anaesthesiol syndrome in an adolescent boy with cerebral palsy. Pediatr Emerg Care 2008;
Scand 1997;41:1033–1038. 24:691–693.
235. Sjoberg M, Karlsson PA, Nordborg C et al. Neuropathologic findings after long- 265. D’Aleo G, Cammaroto S, Rifici C et al. Hallucinations after abrupt withdrawal of
term intrathecal infusion of morphine and bupivacaine for pain treatment in can- oral and intrathecal baclofen. Funct Neurol 2007;22:81–88.
cer patients. Anesthesiology 1992;76:173–186. 266. Ross JC, Cook AM, Stewart GL, Fahy BG. Acute intrathecal baclofen withdrawal: a
236. Ortner CM, Posch M, Roessler B et al. On the ropivacaine-reducing effect of low- brief review of treatment options. Neurocrit Care 2011;14:103–108.
dose sufentanil in intrathecal labor analgesia. Acta Anaesthesiol Scand 2010;54: 267. Fernandes P, Dolan L, Weinstein SL. Intrathecal baclofen withdrawal syndrome fol-
1000–1006. lowing posterior spinal fusion for neuromuscular scoliosis: a case report. Iowa
237. Tejwani GA, Rattan AK, McDonald JS. Role of spinal opioid receptors in the antino- Orthop J 2008;28:77–80.
ciceptive interactions between intrathecal morphine and bupivacaine. Anesth 268. Duhon BS, MacDonald JD. Infusion of intrathecal baclofen for acute withdrawal.
Analg 1992;74:726–734. Technical note. J Neurosurg 2007;107:878–880.
238. Parpaglioni R, Baldassini B, Barbati G, Celleno D. Adding sufentanil to levobupiva- 269. Dalton C, Keenan E, Stevenson V. A novel cause of intrathecal baclofen overdos-
caine or ropivacaine intrathecal anaesthesia affects the minimum local anaesthetic age: lessons to be learnt. Clin Rehabil 2008;22:188–190.
dose required. Acta Anaesthesiol Scand 2009;53:1214–1220. 270. Mercadante S, Intravaia G, Villari P et al. Intrathecal treatment in cancer
239. Levin A, Datta S, Camann WR. Intrathecal ropivacaine for labor analgesia: a com- patients unresponsive to multiple trials of systemic opioids. Clin J Pain 2007;
parison with bupivacaine. Anesth Analg 1998;87:624–627. 23:793–798.
240. van Dongen RT, Crul BJ, van Egmond J. Intrathecal coadministration of bupiva- 271. Krames E. A history of intraspinal analgesia, a small and personal journey. Neuro-
caine diminishes morphine dose progression during long-term intrathecal infusion modulation 2012;15:172–193.
in cancer patients. Clin J Pain 1999;15:166–172. 272. Raffaeli W, Righetti D, Caminiti A et al. Implantable intrathecal pumps for the treat-
241. Ilias W, le Polain B, Buchser E, Demartini L. Patient-controlled analgesia in chronic ment of noncancer chronic pain in the elderly population: drug dose and clinical
pain patients: experience with a new device designed to be used with implanted efficacy. Neuromodulation 2008;11:33–39.
programmable pumps. Pain Pract 2008;8:164–170. 273. Webster LR, Fakata KL, Charapata S, Fisher R, MineHart M. Open-label, multi-
242. Lundborg C, Dahm P, Nitescu P, Biber B. High intrathecal bupivacaine for severe center study of combined intrathecal morphine and ziconotide: addition of
pain in the head and neck. Acta Anaesthesiol Scand 2009;53:908–913. morphine in patients receiving ziconotide for severe chronic pain. Pain Med
243. Yaksh TL. Pharmacology of spinal adrenergic systems which modulate spinal noci- 2008;9:282–290.
ceptive processing. Pharmacol Biochem Behav 1985;22:845–858. 274. Shields DC, Palma C, Khoo LT, Ferrante FM. Extramedullary intrathecal catheter
244. Guevara-L opez U, Aldrete JA, Covarrubias-G omez A, Hernandez-Pando RE, L opez- granuloma adherent to the conus medullaris presenting as cauda equina syn-
Mu~ noz FJ. Absence of histological changes after the administration of a continu- drome. Anesthesiology 2005;102:1059–1061.
ous intrathecal clonidine in Wistar rats. Pain Pract 2009;9:122–129. 275. PrometraV R II programmable pump. Mt. Olive, NJ: Flowonix Medical Inc., 2012.

245. Yaksh TL, Rathbun M, Jage J, Mirzai TH, Grafe M, Hiles RA. Pharmacology and toxi- 276. MRI Guidelines. http://professional.medtronic.com/pt/neuro/idd/ind/mri-guide-
cology of chronically infused epidural clonidine HCl in dogs. Fundam Appl Toxicol lines/#.Vq5SgjYrIUE.
1994;23:319–335. 277. Rauck R, Deer T, Rosen S et al. Accuracy and efficacy of intrathecal administration
246. Bevacqua BK, Fattouh M, Backonja M. Depression, night terrors, and insomnia of morphine sulfate for treatment of intractable pain using the Prometra program-
associated with long-term intrathecal clonidine therapy. Pain Pract 2007;7:36–38. mable pump. Neuromodulation 2010;13:102–108.
247. Rauck RL, North J, Eisenach JC. Intrathecal clonidine and adenosine: effects on 278. Rauck R, Deer T, Rosen S. Long-term follow-up of a novel implantable programma-
pain and sensory processing in patients with chronic regional pain syndrome. Pain ble infusion pump. Neuromodulation 2013;16:163–167.
2015;156:88–95. 279. Rosen SM, Bromberg TA, Padda G et al. Intrathecal administration of InfumorphV R

248. Engelman E, Marsala C. Efficacy of adding clonidine to intrathecal morphine in vs compounded morphine for treatment of intractable pain using the PrometraV R

acute postoperative pain: meta-analysis. Br J Anesth 2013;110:21–27. programmable pump. Pain Med 2013;14:865–873.
249. Koman G, Alfieri A, Rachinger J et al. Erectile dysfunction as rare side effect in the 280. Medtronic. Product Advisories. http://professional.medtronic.com/pt/neuro/idd/
simultaneous intrathecal application of morphine and clonidine. Pain Physician ind/product-advisories/index.htm?cmpid5URL_Neuro_HCP_iddadvisories_-#.
2012;15:E523–E526. Vet6E7eBIZE.
250. Dupoiron D, Richard H, Chabert-Desnot V et al. In vitro stability of low- 281. Tagen KM, Hsu Y, Zhu DC, Linninger AA. CNS wide simulation of low resistance
130

concentration ziconotide alone or in admixtures in intrathecal pumps. Neuromodu- and drug transport due to spinal microanatomy. J Biomech 2015; http://dx.doiorg/
lation 2014;17:472–482. 10.1016/Jbiomech.205.02.018.

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
INTRATHECAL THERAPY BEST PRACTICES AND GUIDELINES

282. Perruchoud C, Eldabe S, Durrer A et al. Effects of flow rate modifications on REFERENCES
reported analgesia and quality of life in chronic pain patients treated with continu-
ous intrathecal drug therapy. Pain Med 2011;12:571–576.
1. Deer TR, Prager J, Levy R, Rathmell J, Buchser E, Burton A et al. Polya-
283. Heittiarachchi HDM, Hsu Y, Harris TJ, Linninger AA. The effect of pulsatile nalgesic Consensus Conference 2012: recommendations for the
flow on intrathecal drug delivery in the spinal canal. J Biomed Eng 2011;39: management of pain by intrathecal (intraspinal) drug delivery: report
2592–2602.
284. Anderson VC, Burcheil KJ, Cooke B. A prospective randomized trial of intrathecal of an interdisciplinary expert panel. Neuromodulation. 2012 Sep-
injection vs epidural infusion in the selection of patients for continuous intrathecal Oct;15:436–64.
opioid therapy. Neuromodulation 2003;6:142–152. 2. Jazz Pharmaceuticals (2015) Jazz Pharmaceuticals Announces Full
285. Grider JS, Harned ME, Sloan PA. Patient selection and trialing techniques utilizing
low-dose intrathecal morphine for chronic nonmalignant pain: a report of two Year And Fourth Quarter 2014 Financial Results [Press Release]
cases. J Opioid Manag 2010;6:371–376. Retrieved from: http://www.prnewswire.com/news-releases/jazz-
286. Reck T, Change EC, Bechir M et al. Applying a part of the daily dose as boli may pharmaceuticals-announces-full-year-and-fourth-quarter-2014-financial-
improve intrathecal opiod therapy in patients with chronic pain. Neuoromodulation
2016; doi: 10.1111/ner.12391. results-300040639.html
287. Nelson DV, Kennington M, Novy DM, Squitieri P. Psychological selection criteria for 3. Institute of Medicine (US) Committee on Advancing Pain Research,
implantable spinal cord stimulators. Pain Forum 1996;5:93–103.
288. Saulino M, Kim PS, Shaw E. Practical considerations and patient selection for intra-
Care, and Education. Relieving Pain in America: A Blueprint for Trans-
thecal drug delivery in the management of chronic pain. J Pain Res 2014;7:627– forming Prevention, Care, Education, and Research. Washington (DC):
638. National Academies Press (US); 2011. 2, Pain as a Public Health Chal-
289. Schatman ME. Selection criteria for intrathecal opioid therapy: a re-examination of
the “science.” Pract Pain Manag 2005; http://www.practicalpainmanagement.com/
lenge. Available from: http://www.ncbi.nlm.nih.gov/books/NBK92516/
treatments/interventional/pumps/selection-criteria-intrathecal-opioid-therapy- 4. CDC. Wide-ranging online data for epidemiologic research (WONDER).
re-examination-science. Atlanta, GA: CDC, National Center for Health Statistics; 2016. Available
at http://wonder.cdc.gov
COMMENTS
***
This refreshing update to the Polyanalgesic Consensus Conference
statement assembles current evidence in basic science, anatomy, physi- For many years the Polyanalgesic Consensus Conference recommen-
ology, engineering and patient care surrounding targeted drug delivery. dations are an important contribution to the management of intrathecal
Thirty concise and tangible recommendations are accompanied by use- therapy. Again, the new version has been thoroughly revised and
ful flow charts, reference tables and decision trees providing guidance includes all relevant features of intrathecal therapy. Of course some
which can be considered for contemporary pain care in patients with points have to remain unanswered and one issue that can always be
targeted drug delivery systems. argued on is the "lines" of intrathecal drugs to choose, especially since
The intrathecal pharmacopeia has expanded in this iteration with divi- there is an almost infinite number of possible combinations. In the end,
sion of first line agents into 1A and 1B categories, and drug choices are every physician has to make an individual choice based on pain- and
now categorized by localized or diffuse pain conditions, as well as cancer patient characteristics, and a recommendation can only be somewhat
or non-cancer related diagnosis. Most notably, the current version largely of a blueprint.
combines the previous neuropathic/nociceptive divisions presented in
the 2012 consensus conference (1), instead focusing on anatomic loca- Tim Reck, MD
tion: diffuse or localized pain in patients with noncancer related pain. Nottwil, Switzerland
Some guidance is sure to prove controversial; while, at times, other ***
recommendations seem anecdotal. Ziconotide maintains a first line
position in both algorithms, yet sales of ziconotide demonstrate a The Polyanalgesic Consensus Conference recommendations on intra-
shrinking year-over-year minority of real-world use (2), and may continue thecal drug delivery is a living document that continues to impact clini-
to decrease with increasing attention on healthcare cost containment. cal practice and guide future research. The fifth iteration of these
Sufentanil monotherapy is recommended for cancer and other terminal recommendations are patient-centered and crafted in a manner that
pain conditions, but there is never a recommendation to use fentanyl condenses the last four years of IDDS research into a practical resource
monotherapy in these same patients. Bupivacaine monotherapy is for practicing clinicians.
placed second line for localized non-cancer pain, but never appears in Critical assessment of evidence from multiple studies are necessary
isolation for the treatment of cancer related pain conditions. At times, when making decisions in day to day practice. Medical literature is
the reader is left wondering: what is anecdote, experience or evidence? immense and not all information is clinically useful. The 2016 guidelines
Amongst the most demanding components of physicianship is the systematically ranked evidence based on the hierarchy of studies. Expert
task of applying a paucity of high-quality clinical evidence to each unique consensus was measured and the degree of recommendations were
patient at the bedside. Regardless of the comprehensiveness of any con- defined. Research gaps were also identified which will hopefully lead to
sensus committee recommendation, clinicians will continue to be scholarly queries and future investigations. I commend the authors for
required to formulate treatment plans in the absence of a randomized their work.
double-blind placebo controlled trial mimicking the patient before them. While appropriate patient selection has been identified as one of the
Perhaps the most notable conclusion upon reading the consensus state- most important factors for a successful outcome when initiating intra-
ment is how few rigorous, well designed clinical studies exist to support thecal therapy, long term management of the intrathecal device poses
clinical practice. With a growing number of patients suffering with chron- several challenges. Pain patterns may change due to an ongoing disease
ic pain (3), and an observed quadrupling of deaths associated with oral process or changes in psychological status years later. Non-medical
opioids (4), the time is ripe for interventional pain physicians to demand determinants of health, such as social and economic status, may strain
funding of high-quality studies to support contemporary pain care. the delivery of needed services to maintain an intrathecal device.
Guidance regarding long term management of the intrathecal device
Robert Bolash, MD was briefly addressed in this manuscript, but more detailed recommen-
131

Cleveland, OH, USA dations would be useful. For example, at what point should a clinician

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132
DEER ET AL.

consider discontinuation of intrathecal therapy because of lack or loss of is probably even more complex than the initial identification of an
efficacy? Is there a consensus on the timing of intrathecal therapy rota- appropriate candidate. Successful outcomes rely upon discovering the
tion from one opioid to another or from an opioid to Ziconotide or to science behind the art of managing the complexities of a long term
combination therapy? Is there consensus on the benefit or lack there of intrathecal pump.
for trailing a different intrathecal medication before rotation? Many of us Brian Bruel, MD, MBA
have been faced with these decisions while in the trenches of clinical Houston, TX, USA
practice. We want to salvage this therapy which was at one point effica-
Comments not included in the Early View version of this paper.
cious, but never ask ourselves when is enough really enough? For many
clinicians with large intrathecal pump practices, long term management
132

www.neuromodulationjournal.com C 2017 International Neuromodulation Society


V Neuromodulation 2017; 20: 96–132

You might also like